1
|
Nturubika BDD, Logan J, Johnson IRD, Moore C, Li KL, Tang J, Lam G, Parkinson-Lawrence E, Williams DB, Chakiris J, Hindes M, Brooks RD, Miles MA, Selemidis S, Gregory P, Weigert R, Butler L, Ward MP, Waugh DJJ, O’Leary JJ, Brooks DA. Components of the Endosome-Lysosome Vesicular Machinery as Drivers of the Metastatic Cascade in Prostate Cancer. Cancers (Basel) 2024; 17:43. [PMID: 39796673 PMCID: PMC11718918 DOI: 10.3390/cancers17010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Prostate cancer remains a significant global health concern, with over 1.4 million new cases diagnosed and more than 330,000 deaths each year. The primary clinical challenge that contributes to poor patient outcomes involves the failure to accurately predict and treat at the onset of metastasis, which remains an incurable stage of the disease. This review discusses the emerging paradigm that prostate cancer metastasis is driven by a dysregulation of critical molecular machinery that regulates endosome-lysosome homeostasis. Endosome and lysosome compartments have crucial roles in maintaining normal cellular function but are also involved in many hallmarks of cancer pathogenesis, including inflammation, immune response, nutrient sensing, metabolism, proliferation, signalling, and migration. Here we discuss new insight into how alterations in the complex network of trafficking machinery, responsible for the microtubule-based transport of endosomes and lysosomes, may be involved in prostate cancer progression. A better understanding of endosome-lysosome dynamics may facilitate the discovery of novel strategies to detect and manage prostate cancer metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Bukuru Dieu-Donne Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Jessica Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Ian R. D. Johnson
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Courtney Moore
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Ka Lok Li
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Jingying Tang
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Giang Lam
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia; (G.L.); (P.G.); (D.J.J.W.)
| | - Emma Parkinson-Lawrence
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Desmond B. Williams
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - James Chakiris
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Madison Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Robert D. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
| | - Mark A. Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (S.S.)
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (S.S.)
| | - Philip Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia; (G.L.); (P.G.); (D.J.J.W.)
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Lisa Butler
- South Australian ImmunoGENomics Cancer Institute, Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA 5000, Australia;
- Solid Tumour Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Mark P. Ward
- Department of Pathology, The Coombe Women and Infants University Hospital, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| | - David J. J. Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia; (G.L.); (P.G.); (D.J.J.W.)
| | - John J. O’Leary
- Department of Histopathology, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| | - Douglas A. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (J.L.); (C.M.); (K.L.L.); (J.T.); (E.P.-L.); (D.B.W.); (J.C.); (M.H.); (R.D.B.)
- Department of Histopathology, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| |
Collapse
|
2
|
de Jonge JJ, Graw A, Kargas V, Batters C, Montanarella AF, O'Loughlin T, Johnson C, Arden SD, Warren AJ, Geeves MA, Kendrick-Jones J, Zaccai NR, Kröss M, Veigel C, Buss F. Motor domain phosphorylation increases nucleotide exchange and turns MYO6 into a faster and stronger motor. Nat Commun 2024; 15:6716. [PMID: 39112473 PMCID: PMC11306250 DOI: 10.1038/s41467-024-49898-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/20/2024] [Indexed: 08/10/2024] Open
Abstract
Myosin motors perform many fundamental functions in eukaryotic cells by providing force generation, transport or tethering capacity. Motor activity control within the cell involves on/off switches, however, few examples are known of how myosins regulate speed or processivity and fine-tune their activity to a specific cellular task. Here, we describe a phosphorylation event for myosins of class VI (MYO6) in the motor domain, which accelerates its ATPase activity leading to a 4-fold increase in motor speed determined by actin-gliding assays, single molecule mechanics and stopped flow kinetics. We demonstrate that the serine/threonine kinase DYRK2 phosphorylates MYO6 at S267 in vitro. Single-molecule optical-tweezers studies at low load reveal that S267-phosphorylation results in faster nucleotide-exchange kinetics without change in the working stroke of the motor. The selective increase in stiffness of the acto-MYO6 complex when proceeding load-dependently into the nucleotide-free rigor state demonstrates that S267-phosphorylation turns MYO6 into a stronger motor. Finally, molecular dynamic simulations of the nucleotide-free motor reveal an alternative interaction network within insert-1 upon phosphorylation, suggesting a molecular mechanism, which regulates insert-1 positioning, turning the S267-phosphorylated MYO6 into a faster motor.
Collapse
Affiliation(s)
- Janeska J de Jonge
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Andreas Graw
- Department of Cellular Physiology, Biomedical Centre (BMC), Ludwig-Maximilians-Universität München, Grosshadernerstrasse 9, 82152, Planegg-Martinsried, Germany
- Centre for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Schellingstrasse 4, 80799, München, Germany
| | - Vasileios Kargas
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Christopher Batters
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
- Department of Cellular Physiology, Biomedical Centre (BMC), Ludwig-Maximilians-Universität München, Grosshadernerstrasse 9, 82152, Planegg-Martinsried, Germany
- Centre for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Schellingstrasse 4, 80799, München, Germany
| | - Antonino F Montanarella
- Department of Cellular Physiology, Biomedical Centre (BMC), Ludwig-Maximilians-Universität München, Grosshadernerstrasse 9, 82152, Planegg-Martinsried, Germany
- Centre for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Schellingstrasse 4, 80799, München, Germany
| | - Tom O'Loughlin
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Chloe Johnson
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Susan D Arden
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Alan J Warren
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | - John Kendrick-Jones
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Nathan R Zaccai
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK
| | - Markus Kröss
- Department of Cellular Physiology, Biomedical Centre (BMC), Ludwig-Maximilians-Universität München, Grosshadernerstrasse 9, 82152, Planegg-Martinsried, Germany
- Centre for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Schellingstrasse 4, 80799, München, Germany
| | - Claudia Veigel
- Department of Cellular Physiology, Biomedical Centre (BMC), Ludwig-Maximilians-Universität München, Grosshadernerstrasse 9, 82152, Planegg-Martinsried, Germany.
- Centre for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Schellingstrasse 4, 80799, München, Germany.
| | - Folma Buss
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
3
|
Behbehani R, Johnson C, Holmes AJ, Gratian MJ, Mulvihill DP, Buss F. The two C. elegans class VI myosins, SPE-15/HUM-3 and HUM-8, share similar motor properties, but have distinct developmental and tissue expression patterns. Front Physiol 2024; 15:1368054. [PMID: 38660538 PMCID: PMC11040104 DOI: 10.3389/fphys.2024.1368054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Myosins of class VI move toward the minus-end of actin filaments and play vital roles in cellular processes such as endocytosis, autophagy, protein secretion, and the regulation of actin filament dynamics. In contrast to the majority of metazoan organisms examined to date which contain a single MYO6 gene, C. elegans, possesses two MYO6 homologues, SPE-15/HUM-3 and HUM-8. Through a combination of in vitro biochemical/biophysical analysis and cellular assays, we confirmed that both SPE-15/HUM-3 and HUM-8 exhibit reverse directionality, velocities, and ATPase activity similar to human MYO6. Our characterization also revealed that unlike SPE-15/HUM-3, HUM-8 is expressed as two distinct splice isoforms, one with an additional unique 14 amino acid insert in the cargo-binding domain. While lipid and adaptor binding sites are conserved in SPE-15/HUM-3 and HUM-8, this conservation does not enable recruitment to endosomes in mammalian cells. Finally, we performed super-resolution confocal imaging on transgenic worms expressing either mNeonGreen SPE-15/HUM-3 or wrmScarlet HUM-8. Our results show a clear distinction in tissue distribution between SPE-15/HUM-3 and HUM-8. While SPE-15/HUM-3 exhibited specific expression in the gonads and neuronal tissue in the head, HUM-8 was exclusively localized in the intestinal epithelium. Overall, these findings align with the established tissue distributions and localizations of human MYO6.
Collapse
Affiliation(s)
- Ranya Behbehani
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Chloe Johnson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Alexander J. Holmes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Matthew J. Gratian
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Niu F, Li L, Wang L, Xiao J, Xu S, Liu Y, Lin L, Yu C, Wei Z. Autoinhibition and activation of myosin VI revealed by its cryo-EM structure. Nat Commun 2024; 15:1187. [PMID: 38331992 PMCID: PMC10853514 DOI: 10.1038/s41467-024-45424-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Myosin VI is the only molecular motor that moves towards the minus end along actin filaments. Numerous cellular processes require myosin VI and tight regulations of the motor's activity. Defects in myosin VI activity are known to cause genetic diseases such as deafness and cardiomyopathy. However, the molecular mechanisms underlying the activity regulation of myosin VI remain elusive. Here, we determined the high-resolution cryo-electron microscopic structure of myosin VI in its autoinhibited state. Our structure reveals that autoinhibited myosin VI adopts a compact, monomeric conformation via extensive interactions between the head and tail domains, orchestrated by an elongated single-α-helix region resembling a "spine". This autoinhibited structure effectively blocks cargo binding sites and represses the motor's ATPase activity. Certain cargo adaptors such as GIPC can release multiple inhibitory interactions and promote motor activity, pointing to a cargo-mediated activation of the processive motor. Moreover, our structural findings allow rationalization of disease-associated mutations in myosin VI. Beyond the activity regulation mechanisms of myosin VI, our study also sheds lights on how activities of other myosin motors such as myosin VII and X might be regulated.
Collapse
Affiliation(s)
- Fengfeng Niu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
| | - Lingxuan Li
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lei Wang
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinman Xiao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Shun Xu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yong Liu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Leishu Lin
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Zhiyi Wei
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Akefe IO, Saber SH, Matthews B, Venkatesh BG, Gormal RS, Blackmore DG, Alexander S, Sieriecki E, Gambin Y, Bertran-Gonzalez J, Vitale N, Humeau Y, Gaudin A, Ellis SA, Michaels AA, Xue M, Cravatt B, Joensuu M, Wallis TP, Meunier FA. The DDHD2-STXBP1 interaction mediates long-term memory via generation of saturated free fatty acids. EMBO J 2024; 43:533-567. [PMID: 38316990 PMCID: PMC10897203 DOI: 10.1038/s44318-024-00030-7] [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: 05/11/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 02/07/2024] Open
Abstract
The phospholipid and free fatty acid (FFA) composition of neuronal membranes plays a crucial role in learning and memory, but the mechanisms through which neuronal activity affects the brain's lipid landscape remain largely unexplored. The levels of saturated FFAs, particularly of myristic acid (C14:0), strongly increase during neuronal stimulation and memory acquisition, suggesting the involvement of phospholipase A1 (PLA1) activity in synaptic plasticity. Here, we show that genetic ablation of the PLA1 isoform DDHD2 in mice dramatically reduces saturated FFA responses to memory acquisition across the brain. Furthermore, DDHD2 loss also decreases memory performance in reward-based learning and spatial memory models prior to the development of neuromuscular deficits that mirror human spastic paraplegia. Via pulldown-mass spectrometry analyses, we find that DDHD2 binds to the key synaptic protein STXBP1. Using STXBP1/2 knockout neurosecretory cells and a haploinsufficient STXBP1+/- mouse model of human early infantile encephalopathy associated with intellectual disability and motor dysfunction, we show that STXBP1 controls targeting of DDHD2 to the plasma membrane and generation of saturated FFAs in the brain. These findings suggest key roles for DDHD2 and STXBP1 in lipid metabolism and in the processes of synaptic plasticity, learning, and memory.
Collapse
Affiliation(s)
- Isaac O Akefe
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
- Academy for Medical Education, Medical School, The University of Queensland, 288 Herston Road, 4006, Brisbane, QLD, Australia
| | - Saber H Saber
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, St Lucia, QLD, 4072, Australia
| | - Benjamin Matthews
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bharat G Venkatesh
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Daniel G Blackmore
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Suzy Alexander
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Emma Sieriecki
- School of Medical Science, University of New South Wales, Randwick, NSW, 2052, Australia
- EMBL Australia, Single Molecule Node, University of New South Wales, Sydney, 2052, Australia
| | - Yann Gambin
- School of Medical Science, University of New South Wales, Randwick, NSW, 2052, Australia
- EMBL Australia, Single Molecule Node, University of New South Wales, Sydney, 2052, Australia
| | | | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, UPR-3212 CNRS - Université de Strasbourg, Strasbourg, France
| | - Yann Humeau
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Université de Bordeaux, Bordeaux, France
| | - Arnaud Gaudin
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Sevannah A Ellis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Alysee A Michaels
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Benjamin Cravatt
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, St Lucia, QLD, 4072, Australia.
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
- The School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
6
|
Gormal RS, Martinez-Marmol R, Brooks AJ, Meunier FA. Location, location, location: Protein kinase nanoclustering for optimised signalling output. eLife 2024; 13:e93902. [PMID: 38206309 PMCID: PMC10783869 DOI: 10.7554/elife.93902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Protein kinases (PKs) are proteins at the core of cellular signalling and are thereby responsible for most cellular physiological processes and their regulations. As for all intracellular proteins, PKs are subjected to Brownian thermal energy that tends to homogenise their distribution throughout the volume of the cell. To access their substrates and perform their critical functions, PK localisation is therefore tightly regulated in space and time, relying upon a range of clustering mechanisms. These include post-translational modifications, protein-protein and protein-lipid interactions, as well as liquid-liquid phase separation, allowing spatial restriction and ultimately regulating access to their substrates. In this review, we will focus on key mechanisms mediating PK nanoclustering in physiological and pathophysiological processes. We propose that PK nanoclusters act as a cellular quantal unit of signalling output capable of integration and regulation in space and time. We will specifically outline the various super-resolution microscopy approaches currently used to elucidate the composition and mechanisms driving PK nanoscale clustering and explore the pathological consequences of altered kinase clustering in the context of neurodegenerative disorders, inflammation, and cancer.
Collapse
Affiliation(s)
- Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbaneAustralia
| | - Ramon Martinez-Marmol
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbaneAustralia
| | - Andrew J Brooks
- Frazer Institute, The University of QueenslandWoolloongabbaAustralia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbaneAustralia
- School of Biomedical Sciences, The University of QueenslandSt LuciaAustralia
| |
Collapse
|
7
|
Skeffington A, Fischer A, Sviben S, Brzezinka M, Górka M, Bertinetti L, Woehle C, Huettel B, Graf A, Scheffel A. A joint proteomic and genomic investigation provides insights into the mechanism of calcification in coccolithophores. Nat Commun 2023; 14:3749. [PMID: 37353496 PMCID: PMC10290126 DOI: 10.1038/s41467-023-39336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 06/05/2023] [Indexed: 06/25/2023] Open
Abstract
Coccolithophores are globally abundant, calcifying microalgae that have profound effects on marine biogeochemical cycles, the climate, and life in the oceans. They are characterized by a cell wall of CaCO3 scales called coccoliths, which may contribute to their ecological success. The intricate morphologies of coccoliths are of interest for biomimetic materials synthesis. Despite the global impact of coccolithophore calcification, we know little about the molecular machinery underpinning coccolithophore biology. Working on the model Emiliania huxleyi, a globally distributed bloom-former, we deploy a range of proteomic strategies to identify coccolithogenesis-related proteins. These analyses are supported by a new genome, with gene models derived from long-read transcriptome sequencing, which revealed many novel proteins specific to the calcifying haptophytes. Our experiments provide insights into proteins involved in various aspects of coccolithogenesis. Our improved genome, complemented with transcriptomic and proteomic data, constitutes a new resource for investigating fundamental aspects of coccolithophore biology.
Collapse
Affiliation(s)
- Alastair Skeffington
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
- Biological and Environmental Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Axel Fischer
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
| | - Sanja Sviben
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
| | - Magdalena Brzezinka
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
| | - Michał Górka
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
| | - Luca Bertinetti
- Max Planck Institute of Colloids and Interfaces, Potsdam-Golm, 14476, Germany
| | - Christian Woehle
- Max Planck Institute for Plant Breeding Research, Max Planck-Genome-Centre Cologne, Cologne, 50829, Germany
| | - Bruno Huettel
- Max Planck Institute for Plant Breeding Research, Max Planck-Genome-Centre Cologne, Cologne, 50829, Germany
| | - Alexander Graf
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany
| | - André Scheffel
- Technische Universität Dresden, Faculty of Biology, 01307, Dresden, Germany.
- Max-Planck Institute of Molecular Plant Physiology, Potsdam-Golm, 14476, Germany.
| |
Collapse
|
8
|
Fyn nanoclustering requires switching to an open conformation and is enhanced by FTLD-Tau biomolecular condensates. Mol Psychiatry 2023; 28:946-962. [PMID: 36258016 PMCID: PMC9908554 DOI: 10.1038/s41380-022-01825-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/08/2022]
Abstract
Fyn is a Src kinase that controls critical signalling cascades and has been implicated in learning and memory. Postsynaptic enrichment of Fyn underpins synaptotoxicity in dementias such as Alzheimer's disease and frontotemporal lobar degeneration with Tau pathology (FTLD-Tau). The FLTD P301L mutant Tau is associated with a higher propensity to undergo liquid-liquid phase separation (LLPS) and form biomolecular condensates. Expression of P301L mutant Tau promotes aberrant trapping of Fyn in nanoclusters within hippocampal dendrites by an unknown mechanism. Here, we used single-particle tracking photoactivated localisation microscopy to demonstrate that the opening of Fyn into its primed conformation promotes its nanoclustering in dendrites leading to increased Fyn/ERK/S6 downstream signalling. Preventing the auto-inhibitory closed conformation of Fyn through phospho-inhibition or through perturbation of its SH3 domain increased Fyn's nanoscale trapping, whereas inhibition of the catalytic domain had no impact. By combining pharmacological and genetic approaches, we demonstrate that P301L Tau enhanced both Fyn nanoclustering and Fyn/ERK/S6 signalling via its ability to form biomolecular condensates. Together, our findings demonstrate that Fyn alternates between a closed and an open conformation, the latter being enzymatically active and clustered. Furthermore, pathogenic immobilisation of Fyn relies on the ability of P301L Tau to form biomolecular condensates, thus highlighting the critical importance of LLPS in controlling nanoclustering and downstream intracellular signalling events.
Collapse
|
9
|
Schenk EB, Meunier FA, Oelz DB. Spatial redistribution of neurosecretory vesicles upon stimulation accelerates their directed transport to the plasma membrane. PLoS One 2022; 17:e0264521. [PMID: 35294476 PMCID: PMC8926195 DOI: 10.1371/journal.pone.0264521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/11/2022] [Indexed: 11/19/2022] Open
Abstract
Through the integration of results from an imaging analysis of intracellular trafficking of labelled neurosecretory vesicles in chromaffin cells, we develop a Markov state model to describe their transport and binding kinetics. Our simulation results indicate that a spatial redistribution of neurosecretory vesicles occurs upon secretagogue stimulation leading vesicles to the plasma membrane where they undergo fusion thereby releasing adrenaline and noradrenaline. Furthermore, we find that this redistribution alone can explain the observed up-regulation of vesicle transport upon stimulation and its directional bias towards the plasma membrane. Parameter fitting indicates that in the deeper compartment within the cell, vesicle transport is asymmetric and characterised by a bias towards the plasma membrane.
Collapse
Affiliation(s)
- Elaine B. Schenk
- School of Mathematics & Physics, The University of Queensland, Brisbane, Australia
| | - Frederic A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute (QBI), The University of Queensland, Brisbane, Australia
| | - Dietmar B. Oelz
- School of Mathematics & Physics, The University of Queensland, Brisbane, Australia
- * E-mail:
| |
Collapse
|
10
|
Zakrzewski P, Lenartowska M, Buss F. Diverse functions of myosin VI in spermiogenesis. Histochem Cell Biol 2021; 155:323-340. [PMID: 33386429 PMCID: PMC8021524 DOI: 10.1007/s00418-020-01954-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Spermiogenesis is the final stage of spermatogenesis, a differentiation process during which unpolarized spermatids undergo excessive remodeling that results in the formation of sperm. The actin cytoskeleton and associated actin-binding proteins play crucial roles during this process regulating organelle or vesicle delivery/segregation and forming unique testicular structures involved in spermatid remodeling. In addition, several myosin motor proteins including MYO6 generate force and movement during sperm differentiation. MYO6 is highly unusual as it moves towards the minus end of actin filaments in the opposite direction to other myosin motors. This specialized feature of MYO6 may explain the many proposed functions of this myosin in a wide array of cellular processes in animal cells, including endocytosis, secretion, stabilization of the Golgi complex, and regulation of actin dynamics. These diverse roles of MYO6 are mediated by a range of specialized cargo-adaptor proteins that link this myosin to distinct cellular compartments and processes. During sperm development in a number of different organisms, MYO6 carries out pivotal functions. In Drosophila, the MYO6 ortholog regulates actin reorganization during spermatid individualization and male KO flies are sterile. In C. elegans, the MYO6 ortholog mediates asymmetric segregation of cytosolic material and spermatid budding through cytokinesis, whereas in mice, this myosin regulates assembly of highly specialized actin-rich structures and formation of membrane compartments to allow the formation of fully differentiated sperm. In this review, we will present an overview and compare the diverse function of MYO6 in the specialized adaptations of spermiogenesis in flies, worms, and mammals.
Collapse
Affiliation(s)
- Przemysław Zakrzewski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Marta Lenartowska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
11
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
12
|
Abstract
Unconventional myosins are a large superfamily of actin-based molecular motors that use ATP as fuel to generate mechanical motions/forces. The distinct tails in different unconventional myosin subfamilies can recognize various cargoes including proteins and lipids. Thus, they can play diverse roles in many biological processes such as cellular trafficking, mechanical supports, force sensing, etc. This chapter focuses on some recent advances on the structural studies of how unconventional myosins specifically bind to cargoes with their cargo-binding domains.
Collapse
|
13
|
Zakrzewski P, Rędowicz MJ, Buss F, Lenartowska M. Loss of myosin VI expression affects acrosome/acroplaxome complex morphology during mouse spermiogenesis†. Biol Reprod 2020; 103:521-533. [PMID: 32412041 PMCID: PMC7442776 DOI: 10.1093/biolre/ioaa071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/24/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
During spermiogenesis in mammals, actin filaments and a variety of actin-binding proteins are involved in the formation and function of highly specialized testis-specific structures. Actin-based motor proteins, such as myosin Va and VIIa, play a key role in this complex process of spermatid transformation into mature sperm. We have previously demonstrated that myosin VI (MYO6) is also expressed in mouse testes. It is present in actin-rich structures important for spermatid development, including one of the earliest events in spermiogenesis—acrosome formation. Here, we demonstrate using immunofluorescence, cytochemical, and ultrastructural approaches that MYO6 is involved in maintaining the structural integrity of these specialized actin-rich structures during acrosome biogenesis in mouse. We show that MYO6 together with its binding partner TOM1/L2 is present at/around the spermatid Golgi complex and the nascent acrosome. Depletion of MYO6 in Snell’s waltzer mice causes structural disruptions of the Golgi complex and affects the acrosomal granule positioning within the developing acrosome. In summary, our results suggest that MYO6 plays an anchoring role during the acrosome biogenesis mainly by tethering of different cargo/membranes to highly specialized actin-related structures.
Collapse
Affiliation(s)
- Przemysław Zakrzewski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Cambridge, UK
| | - Marta Lenartowska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| |
Collapse
|
14
|
O'Loughlin T, Kendrick-Jones J, Buss F. Approaches to Identify and Characterise MYO6-Cargo Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:355-380. [PMID: 32451866 DOI: 10.1007/978-3-030-38062-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the prevalence and importance of the actin cytoskeleton and the host of associated myosin motors, it comes as no surprise to find that they are linked to a plethora of cellular functions and pathologies. Although our understanding of the biophysical properties of myosin motors has been aided by the high levels of conservation in their motor domains and the extensive work on myosin in skeletal muscle contraction, our understanding of how the nonmuscle myosins participate in such a wide variety of cellular processes is less clear. It is now well established that the highly variable myosin tails are responsible for targeting these myosins to distinct cellular sites for specific functions, and although a number of adaptor proteins have been identified, our current understanding of the cellular processes involved is rather limited. Furthermore, as more adaptor proteins, cargoes and complexes are identified, the importance of elucidating the regulatory mechanisms involved is essential. Ca2+, and now phosphorylation and ubiquitination, are emerging as important regulators of cargo binding, and it is likely that other post-translational modifications are also involved. In the case of myosin VI (MYO6), a number of immediate binding partners have been identified using traditional approaches such as yeast two-hybrid screens and affinity-based pull-downs. However, these methods have only been successful in identifying the cargo adaptors, but not the cargoes themselves, which may often comprise multi-protein complexes. Furthermore, motor-adaptor-cargo interactions are dynamic by nature and often weak, transient and highly regulated and therefore difficult to capture using traditional affinity-based methods. In this chapter we will discuss the various approaches including functional proteomics that have been used to uncover and characterise novel MYO6-associated proteins and complexes and how this work contributes to a fuller understanding of the targeting and function(s) of this unique myosin motor.
Collapse
Affiliation(s)
- Thomas O'Loughlin
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK.
| |
Collapse
|
15
|
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: 30] [Impact Index Per Article: 5.0] [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
|
16
|
Carbone E, Borges R, Eiden LE, García AG, Hernández-Cruz A. Chromaffin Cells of the Adrenal Medulla: Physiology, Pharmacology, and Disease. Compr Physiol 2019; 9:1443-1502. [PMID: 31688964 DOI: 10.1002/cphy.c190003] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Chromaffin cells (CCs) of the adrenal gland and the sympathetic nervous system produce the catecholamines (epinephrine and norepinephrine; EPI and NE) needed to coordinate the bodily "fight-or-flight" response to fear, stress, exercise, or conflict. EPI and NE release from CCs is regulated both neurogenically by splanchnic nerve fibers and nonneurogenically by hormones (histamine, corticosteroids, angiotensin, and others) and paracrine messengers [EPI, NE, adenosine triphosphate, opioids, γ-aminobutyric acid (GABA), etc.]. The "stimulus-secretion" coupling of CCs is a Ca2+ -dependent process regulated by Ca2+ entry through voltage-gated Ca2+ channels, Ca2+ pumps, and exchangers and intracellular organelles (RE and mitochondria) and diffusible buffers that provide both Ca2+ -homeostasis and Ca2+ -signaling that ultimately trigger exocytosis. CCs also express Na+ and K+ channels and ionotropic (nAChR and GABAA ) and metabotropic receptors (mACh, PACAP, β-AR, 5-HT, histamine, angiotensin, and others) that make CCs excitable and responsive to autocrine and paracrine stimuli. To maintain high rates of E/NE secretion during stressful conditions, CCs possess a large number of secretory chromaffin granules (CGs) and members of the soluble NSF-attachment receptor complex protein family that allow docking, fusion, and exocytosis of CGs at the cell membrane, and their recycling. This article attempts to provide an updated account of well-established features of the molecular processes regulating CC function, and a survey of the as-yet-unsolved but important questions relating to CC function and dysfunction that have been the subject of intense research over the past 15 years. Examples of CCs as a model system to understand the molecular mechanisms associated with neurodegenerative diseases are also provided. Published 2019. Compr Physiol 9:1443-1502, 2019.
Collapse
Affiliation(s)
- Emilio Carbone
- Laboratory of Cellular and Molecular Neuroscience, Department of Drug Science, N.I.S. Centre, University of Torino, Torino, Italy
| | - Ricardo Borges
- Unidad de Farmacología, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Antonio G García
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
| | - Arturo Hernández-Cruz
- Departamento de Neurociencia Cognitiva and Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autonoma de México, Ciudad Universitaria, CDMX, México
| |
Collapse
|
17
|
de Jonge JJ, Batters C, O'Loughlin T, Arden SD, Buss F. The MYO6 interactome: selective motor-cargo complexes for diverse cellular processes. FEBS Lett 2019; 593:1494-1507. [PMID: 31206648 DOI: 10.1002/1873-3468.13486] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022]
Abstract
Myosins of class VI (MYO6) are unique actin-based motor proteins that move cargo towards the minus ends of actin filaments. As the sole myosin with this directionality, it is critically important in a number of biological processes. Indeed, loss or overexpression of MYO6 in humans is linked to a variety of pathologies including deafness, cardiomyopathy, neurodegenerative diseases as well as cancer. This myosin interacts with a wide variety of direct binding partners such as for example the selective autophagy receptors optineurin, TAX1BP1 and NDP52 and also Dab2, GIPC, TOM1 and LMTK2, which mediate distinct functions of different MYO6 isoforms along the endocytic pathway. Functional proteomics has recently been used to identify the wider MYO6 interactome including several large functionally distinct multi-protein complexes, which highlight the importance of this myosin in regulating the actin and septin cytoskeleton. Interestingly, adaptor-binding not only triggers cargo attachment, but also controls the inactive folded conformation and dimerisation of MYO6. Thus, the C-terminal tail domain mediates cargo recognition and binding, but is also crucial for modulating motor activity and regulating cytoskeletal track dynamics.
Collapse
Affiliation(s)
| | | | - Thomas O'Loughlin
- Cambridge Institute for Medical Research, University of Cambridge, UK
| | - Susan D Arden
- Cambridge Institute for Medical Research, University of Cambridge, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, UK
| |
Collapse
|
18
|
Wagner W, Lippmann K, Heisler FF, Gromova KV, Lombino FL, Roesler MK, Pechmann Y, Hornig S, Schweizer M, Polo S, Schwarz JR, Eilers J, Kneussel M. Myosin VI Drives Clathrin-Mediated AMPA Receptor Endocytosis to Facilitate Cerebellar Long-Term Depression. Cell Rep 2019; 28:11-20.e9. [DOI: 10.1016/j.celrep.2019.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 05/01/2019] [Accepted: 05/31/2019] [Indexed: 11/30/2022] Open
|
19
|
Deng H, Dutta P, Liu J. Stochastic modeling of nanoparticle internalization and expulsion through receptor-mediated transcytosis. NANOSCALE 2019; 11:11227-11235. [PMID: 31157808 PMCID: PMC6634982 DOI: 10.1039/c9nr02710f] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Receptor-mediated transcytosis (RMT) is a fundamental mechanism for the transcellular transport of nanoparticles. RMT is a complex process, during which the nanoparticles actively interact with the membrane and the membrane profile undergoes extreme deformations for particle internalization and expulsion. In this work, we developed a stochastic model to study the endocytosis and exocytosis of nanoparticles across soft membranes. The model is based on the combination of a stochastic particle binding model with a membrane model, and accounts for both clathrin-mediated endocytosis for internalization and actin-mediated exocytosis for expulsion. Our results showed that nanoparticles must have certain avidity with enough ligand density and ligand-receptor binding affinity to be taken up, while too high avidity limited the particle release from the cell surface. We further explored the functional roles of actin during exocytosis, which has been a topic under active debate. Our simulations indicated that the membrane compression due to the actin induced tension tended to break the ligand-receptor bonds and to shrink the fusion pore. Therefore, an intermediate tension promoted the fusion pore expansion and nanoparticle release, while high tension prohibits particle release. Our model provides new and critical mechanistic insights into RMT, and represents a powerful platform for aiding the rational design of nanocarriers for controlled drug delivery.
Collapse
Affiliation(s)
- Hua Deng
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99163, USA.
| | - Prashanta Dutta
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99163, USA.
| | - Jin Liu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99163, USA.
| |
Collapse
|
20
|
Zhang X, Han L, Wang Q, Zhang C, Yu Y, Tian J, Kong Z. The host actin cytoskeleton channels rhizobia release and facilitates symbiosome accommodation during nodulation in Medicago truncatula. THE NEW PHYTOLOGIST 2019; 221:1049-1059. [PMID: 30156704 DOI: 10.1111/nph.15423] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/02/2018] [Indexed: 05/13/2023]
Abstract
In plants, the actin cytoskeleton plays a central role in regulating intracellular transport and trafficking in the endomembrane system. Work in legumes suggested that during nodulation, the actin cytoskeleton coordinates numerous cellular processes in the development of nitrogen-fixing nodules. However, we lacked live-cell visualizations demonstrating dynamic remodeling of the actin cytoskeleton during infection droplet release and symbiosome development. Here, we generated transgenic Medicago truncatula lines stably expressing the fluorescent actin marker ABD2-GFP, and utilized live-cell imaging to reveal the architecture and dynamics of the actin cytoskeleton during nodule development. Live-cell observations showed that different zones in nitrogen-fixing nodules exhibit distinct actin architectures and infected cells display five characteristic actin architectures during nodule development. Live-cell imaging combined with three-dimensional reconstruction demonstrated that dense filamentous-actin (F-actin) arrays channel the elongation of infection threads and the release of infection droplets, an F-actin network encircles freshly-released rhizobia, and short F-actin fragments and actin dots around radially distributed symbiosomes. Our findings suggest an important role of the actin cytoskeleton in infection droplet release, symbiosome development and maturation, and provide significant insight into the cellular mechanisms underlying nodule development and nitrogen fixation during legume-rhizobia interactions.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Libo Han
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Wang
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Zhang
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanjun Yu
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Juan Tian
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhaosheng Kong
- State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
21
|
Gimenez-Molina Y, Villanueva J, Francés MDM, Viniegra S, Gutiérrez LM. Multiple Mechanisms Driving F-actin-Dependent Transport of Organelles to and From Secretory Sites in Bovine Chromaffin Cells. Front Cell Neurosci 2018; 12:344. [PMID: 30356839 PMCID: PMC6190647 DOI: 10.3389/fncel.2018.00344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/18/2018] [Indexed: 01/12/2023] Open
Abstract
Neuroendocrine chromaffin cells represent an excellent model to study the molecular mechanisms associated with the exo-endocytotic cycle of neurotransmitter release. In this study, EGFP-Lifeact and confocal microscopy has been used to analyze the re-organization of the cortical F-actin cytoskeleton associated to organelle transport during secretion with unprecedented detail. In these cells secretory events accumulate in temperature-sensitive and myosin II-dependent F-actin expansions and retractions affecting specific regions of the sub-membrane space. Interestingly, not only vesicles but also mitochondria are transported toward the plasmalemma during these expansions. Simultaneously, we found F-actin cytoskeletal retraction withdraws vesicles from the sub-plasmalemmal space, forming novel empty internal spaces into which organelles can be transported. In addition to these well-coordinated, F-actin-myosin II dependent processes that drive the transport of the majority of vesicles, fast transport of chromaffin vesicles was observed, albeit less frequently, which used F-actin comet tails nucleated from the granular membrane. Thus, upon cell stimulation F-actin structures use diverse mechanisms to transport organelles to and from the membrane during the exo-endocytotic cycle taking place in specific areas of cell periphery.
Collapse
Affiliation(s)
- Yolanda Gimenez-Molina
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - José Villanueva
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Maria Del Mar Francés
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Salvador Viniegra
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Luis M Gutiérrez
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
22
|
Actin Remodeling in Regulated Exocytosis: Toward a Mesoscopic View. Trends Cell Biol 2018; 28:685-697. [DOI: 10.1016/j.tcb.2018.04.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/05/2018] [Accepted: 04/13/2018] [Indexed: 01/10/2023]
|
23
|
Yoshida A, Sakai N, Uekusa Y, Imaoka Y, Itagaki Y, Suzuki Y, Yoshimura SH. Morphological changes of plasma membrane and protein assembly during clathrin-mediated endocytosis. PLoS Biol 2018; 16:e2004786. [PMID: 29723197 PMCID: PMC5953504 DOI: 10.1371/journal.pbio.2004786] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/15/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022] Open
Abstract
Clathrin-mediated endocytosis (CME) proceeds through a series of morphological changes of the plasma membrane induced by a number of protein components. Although the spatiotemporal assembly of these proteins has been elucidated by fluorescence-based techniques, the protein-induced morphological changes of the plasma membrane have not been fully clarified in living cells. Here, we visualize membrane morphology together with protein localizations during CME by utilizing high-speed atomic force microscopy (HS-AFM) combined with a confocal laser scanning unit. The plasma membrane starts to invaginate approximately 30 s after clathrin starts to assemble, and the aperture diameter increases as clathrin accumulates. Actin rapidly accumulates around the pit and induces a small membrane swelling, which, within 30 s, rapidly covers the pit irreversibly. Inhibition of actin turnover abolishes the swelling and induces a reversible open–close motion of the pit, indicating that actin dynamics are necessary for efficient and irreversible pit closure at the end of CME. Cells communicate with their environments via the plasma membrane and various membrane proteins. Clathrin-mediated endocytosis (CME) plays a central role in such communication and proceeds with a series of multiprotein assembly, deformation of the plasma membrane, and production of a membrane vesicle that delivers extracellular signaling molecules into the cytoplasm. In this study, we utilized our home-built correlative imaging system comprising high-speed atomic force microscopy (HS-AFM) and confocal fluorescence microscopy to simultaneously image morphological changes of the plasma membrane and protein localization during CME in a living cell. The results revealed a tight correlation between the size of the pit and the amount of clathrin assembled. Actin dynamics play multiple roles in the assembly, maturation, and closing phases of the process, and affects membrane morphology, suggesting a close relationship between endocytosis and dynamic events at the cell cortex. Knock down of dynamin also affected the closing motion of the pit and showed functional correlation with actin.
Collapse
Affiliation(s)
- Aiko Yoshida
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | | | - Yuka Imaoka
- R&D Group, Olympus Corporation, Hachioji, Japan
| | | | - Yuki Suzuki
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | | |
Collapse
|
24
|
Bademosi AT, Lauwers E, Amor R, Verstreken P, van Swinderen B, Meunier FA. In Vivo Single-Molecule Tracking at the Drosophila Presynaptic Motor Nerve Terminal. J Vis Exp 2018. [PMID: 29364242 PMCID: PMC5908646 DOI: 10.3791/56952] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
An increasing number of super-resolution microscopy techniques are helping to uncover the mechanisms that govern the nanoscale cellular world. Single-molecule imaging is gaining momentum as it provides exceptional access to the visualization of individual molecules in living cells. Here, we describe a technique that we developed to perform single-particle tracking photo-activated localization microscopy (sptPALM) in Drosophila larvae. Synaptic communication relies on key presynaptic proteins that act by docking, priming, and promoting the fusion of neurotransmitter-containing vesicles with the plasma membrane. A range of protein-protein and protein-lipid interactions tightly regulates these processes and the presynaptic proteins therefore exhibit changes in mobility associated with each of these key events. Investigating how mobility of these proteins correlates with their physiological function in an intact live animal is essential to understanding their precise mechanism of action. Extracting protein mobility with high resolution in vivo requires overcoming limitations such as optical transparency, accessibility, and penetration depth. We describe how photoconvertible fluorescent proteins tagged to the presynaptic protein Syntaxin-1A can be visualized via slight oblique illumination and tracked at the motor nerve terminal or along the motor neuron axon of the third instar Drosophila larva.
Collapse
Affiliation(s)
- Adekunle T Bademosi
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland
| | - Elsa Lauwers
- VIB Centre for Brain and Disease Research, KU Leuven Department of Neurosciences, Leuven Institute for Neurodegenerative Disease (LIND)
| | - Rumelo Amor
- Queensland Brain Institute, The University of Queensland
| | - Patrik Verstreken
- VIB Centre for Brain and Disease Research, KU Leuven Department of Neurosciences, Leuven Institute for Neurodegenerative Disease (LIND)
| | | | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland;
| |
Collapse
|
25
|
How does the stimulus define exocytosis in adrenal chromaffin cells? Pflugers Arch 2017; 470:155-167. [DOI: 10.1007/s00424-017-2052-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 12/28/2022]
|
26
|
Tomatis VM, Josh P, Papadopulos A, Gormal RS, Lanoue V, Martin S, Meunier FA. ENA/VASP proteins regulate exocytosis by mediating myosin VI-dependent recruitment of secretory granules to the cortical actin network. Mol Cell Neurosci 2017; 84:100-111. [PMID: 28784263 DOI: 10.1016/j.mcn.2017.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/17/2017] [Accepted: 07/27/2017] [Indexed: 10/24/2022] Open
Abstract
In neurosecretory cells, myosin VI associated with secretory granules (SGs) mediates their activity-dependent recruitment to the cortical actin network and is necessary to sustain exocytosis. The mechanism by which myosin VI interacts with SGs is unknown. Using a myosin VI pull-down assay and mass spectrometry we identified Mena, a member of the ENA/VASP family, as a myosin VI binding partner in PC12 cells, and confirmed that Mena colocalized with myosin VI on SGs. Using a knock-sideways approach to inactivate the ENA/VASP family members by mitochondrial relocation, we revealed a concomitant redistribution of myosin VI. This was ensued by a reduction in the association of myosin VI with SGs, a decreased SG mobility and density in proximity to the plasma membrane as well as decreased evoked exocytosis. These data demonstrate that ENA/VASP proteins regulate SG exocytosis through modulating the activity of myosin VI.
Collapse
Affiliation(s)
- Vanesa M Tomatis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Peter Josh
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andreas Papadopulos
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sally Martin
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
27
|
Gutiérrez LM, Villanueva J. The role of F-actin in the transport and secretion of chromaffin granules: an historic perspective. Pflugers Arch 2017; 470:181-186. [PMID: 28730385 PMCID: PMC5748413 DOI: 10.1007/s00424-017-2040-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/23/2022]
Abstract
Actin is one of the most ubiquitous protein playing fundamental roles in a variety of cellular processes. Since early in the 1980s, it was evident that filamentous actin (F-actin) formed a peripheral cortical barrier that prevented vesicles to access secretory sites in chromaffin cells in culture. Later, around 2000, it was described that the F-actin structure accomplishes a dual role serving both vesicle transport and retentive purposes and undergoing dynamic transient changes during cell stimulation. The complex role of the F-actin cytoskeleton in neuroendocrine secretion was further evidenced when it has been proved to participate in the scaffold structure holding together the secretory machinery at active sites and participate in the generation of mechanical forces that drive the opening of the fusion pore, during the first decade of the present century. The complex vision of the multiple roles of F-actin in secretion we have acquired to date comes largely from studies performed on traditional 2D cultures of primary cells; however, recent evidences suggest that these may not accurately mimic the 3D in vivo environment, and thus, more work is now needed on adrenomedullary cells kept in a more “native” configuration to fully understand the role of F-actin in regulating chromaffin granule transport and secretion under physiological conditions.
Collapse
Affiliation(s)
- Luis M Gutiérrez
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant, 03550, Alicante, Spain.
| | - José Villanueva
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant, 03550, Alicante, Spain
| |
Collapse
|
28
|
Delestre-Delacour C, Carmon O, Laguerre F, Estay-Ahumada C, Courel M, Elias S, Jeandel L, Rayo MV, Peinado JR, Sengmanivong L, Gasman S, Coudrier E, Anouar Y, Montero-Hadjadje M. Myosin 1b and F-actin are involved in the control of secretory granule biogenesis. Sci Rep 2017; 7:5172. [PMID: 28701771 PMCID: PMC5507975 DOI: 10.1038/s41598-017-05617-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/31/2017] [Indexed: 12/22/2022] Open
Abstract
Hormone secretion relies on secretory granules which store hormones in endocrine cells and release them upon cell stimulation. The molecular events leading to hormone sorting and secretory granule formation at the level of the TGN are still elusive. Our proteomic analysis of purified whole secretory granules or secretory granule membranes uncovered their association with the actomyosin components myosin 1b, actin and the actin nucleation complex Arp2/3. We found that myosin 1b controls the formation of secretory granules and the associated regulated secretion in both neuroendocrine cells and chromogranin A-expressing COS7 cells used as a simplified model of induced secretion. We show that F-actin is also involved in secretory granule biogenesis and that myosin 1b cooperates with Arp2/3 to recruit F-actin to the Golgi region where secretory granules bud. These results provide the first evidence that components of the actomyosin complex promote the biogenesis of secretory granules and thereby regulate hormone sorting and secretion.
Collapse
Affiliation(s)
- Charlène Delestre-Delacour
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France
| | - Ophélie Carmon
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France
| | - Fanny Laguerre
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France
| | - Catherine Estay-Ahumada
- Université de Strasbourg, CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, 67000, Strasbourg, France
| | - Maïté Courel
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France.,CNRS-UPMC FRE3402, Pierre et Marie Curie University, 75252, Paris, Cedex 05, France
| | - Salah Elias
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France.,University of Oxford, Sir William Dunn School of Pathology, Oxford, Oxfordshire, United Kingdom
| | - Lydie Jeandel
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France
| | - Margarita Villar Rayo
- Instituto de Investigación en Recursos Cinegéticos, Proteomics Core Facility, 13071, Ciudad Real, Spain
| | - Juan R Peinado
- Laboratory of oxidative stress and neurodegeneration, Facultad de Medicina de Ciudad Real, 13071, Ciudad Real, Spain
| | - Lucie Sengmanivong
- Institut Curie - PSL Research University, Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Nikon Imaging Centre, 75005, Paris, France
| | - Stéphane Gasman
- Université de Strasbourg, CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, 67000, Strasbourg, France
| | - Evelyne Coudrier
- CNRS UMR 144 Cell Signaling and Morphogenesis, Institut Curie, 75005, Paris, France
| | - Youssef Anouar
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France.
| | - Maité Montero-Hadjadje
- Normandie Univ, UNIROUEN, INSERM, U1239, Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie, 76000, Rouen, France.
| |
Collapse
|
29
|
Fan F, Matsunaga K, Wang H, Ishizaki R, Kobayashi E, Kiyonari H, Mukumoto Y, Okunishi K, Izumi T. Exophilin-8 assembles secretory granules for exocytosis in the actin cortex via interaction with RIM-BP2 and myosin-VIIa. eLife 2017; 6. [PMID: 28673385 PMCID: PMC5496739 DOI: 10.7554/elife.26174] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/04/2017] [Indexed: 12/15/2022] Open
Abstract
Exophilin-8 has been reported to play a role in anchoring secretory granules within the actin cortex, due to its direct binding activities to Rab27 on the granule membrane and to F-actin and its motor protein, myosin-Va. Here, we show that exophilin-8 accumulates granules in the cortical F-actin network not by direct interaction with myosin-Va, but by indirect interaction with a specific form of myosin-VIIa through its previously unknown binding partner, RIM-BP2. RIM-BP2 also associates with exocytic machinery, Cav1.3, RIM, and Munc13-1. Disruption of the exophilin-8-RIM-BP2-myosin-VIIa complex by ablation or knockdown of each component markedly decreases both the peripheral accumulation and exocytosis of granules. Furthermore, exophilin-8-null mouse pancreatic islets lose polarized granule localization at the β-cell periphery and exhibit impaired insulin secretion. This newly identified complex acts as a physical and functional scaffold and provides a mechanism supporting a releasable pool of granules within the F-actin network beneath the plasma membrane.
Collapse
Affiliation(s)
- Fushun Fan
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Kohichi Matsunaga
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hao Wang
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Ray Ishizaki
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Eri Kobayashi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hiroshi Kiyonari
- Animal Resource Development Unit, RIKEN Center for Life Science Technologies, Kobe, Japan.,Genetic Engineering Team, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Yoshiko Mukumoto
- Genetic Engineering Team, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Katsuhide Okunishi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.,Research Program for Signal Transduction, Division of Endocrinology, Metabolism and Signal Research, Gunma University Initiative for Advanced Research, Maebashi, Japan
| |
Collapse
|
30
|
Papadopulos A. Membrane shaping by actin and myosin during regulated exocytosis. Mol Cell Neurosci 2017; 84:93-99. [PMID: 28536001 DOI: 10.1016/j.mcn.2017.05.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/21/2017] [Accepted: 05/19/2017] [Indexed: 12/01/2022] Open
Abstract
The cortical actin network in neurosecretory cells is a dense mesh of actin filaments underlying the plasma membrane. Interaction of actomyosin with vesicular membranes or the plasma membrane is vital for tethering, retention, transport as well as fusion and fission of exo- and endocytic membrane structures. During regulated exocytosis the cortical actin network undergoes dramatic changes in morphology to accommodate vesicle docking, fusion and replenishment. Most of these processes involve plasma membrane Phosphoinositides (PIP) and investigating the interactions between the actin cortex and secretory structures has become a hotbed for research in recent years. Actin remodelling leads to filopodia outgrowth and the creation of new fusion sites in neurosecretory cells and actin, myosin and dynamin actively shape and maintain the fusion pore of secretory vesicles. Changes in viscoelastic properties of the actin cortex can facilitate vesicular transport and lead to docking and priming of vesicle at the plasma membrane. Small GTPase actin mediators control the state of the cortical actin network and influence vesicular access to their docking and fusion sites. These changes potentially affect membrane properties such as tension and fluidity as well as the mobility of embedded proteins and could influence the processes leading to both exo- and endocytosis. Here we discuss the multitudes of actin and membrane interactions that control successive steps underpinning regulated exocytosis.
Collapse
Affiliation(s)
- Andreas Papadopulos
- The Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
31
|
Abstract
Myosin VI (MVI) is a versatile actin-based motor protein that has been implicated in a variety of different cellular processes, including endo- and exocytic vesicle trafficking, Golgi morphology, and actin structure stabilization. A role for MVI in crucial actin-based processes involved in sperm maturation was demonstrated in Drosophila. Because of the prominence and importance of actin structures in mammalian spermiogenesis, we investigated whether MVI was associated with actin-mediated maturation events in mammals. Both immunofluorescence and ultrastructural analyses using immunogold labeling showed that MVI was strongly linked with key structures involved in sperm development and maturation. During the early stage of spermiogenesis, MVI is associated with the Golgi and with coated and uncoated vesicles, which fuse to form the acrosome. Later, as the acrosome spreads to form a cap covering the sperm nucleus, MVI is localized to the acroplaxome, an actin-rich structure that anchors the acrosome to the nucleus. Finally, during the elongation/maturation phase, MVI is associated with the actin-rich structures involved in nuclear shaping: the acroplaxome, manchette, and Sertoli cell actin hoops. Since this is the first report of MVI expression and localization during mouse spermiogenesis and MVI partners in developing sperm have not yet been identified, we discuss some probable roles for MVI in this process. During early stages, MVI is hypothesized to play a role in Golgi morphology and function as well as in actin dynamics regulation important for attachment of developing acrosome to the nuclear envelope. Next, the protein might also play anchoring roles to help generate forces needed for spermatid head elongation. Moreover, association of MVI with actin that accumulates in the Sertoli cell ectoplasmic specialization and other actin structures in surrounding cells suggests additional MVI functions in spermatid movement across the seminiferous epithelium and in sperm release.
Collapse
|
32
|
Bademosi AT, Lauwers E, Padmanabhan P, Odierna L, Chai YJ, Papadopulos A, Goodhill GJ, Verstreken P, van Swinderen B, Meunier FA. In vivo single-molecule imaging of syntaxin1A reveals polyphosphoinositide- and activity-dependent trapping in presynaptic nanoclusters. Nat Commun 2017; 8:13660. [PMID: 28045048 PMCID: PMC5171881 DOI: 10.1038/ncomms13660] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/21/2016] [Indexed: 01/03/2023] Open
Abstract
Syntaxin1A is organized in nanoclusters that are critical for the docking and priming of secretory vesicles from neurosecretory cells. Whether and how these nanoclusters are affected by neurotransmitter release in nerve terminals from a living organism is unknown. Here we imaged photoconvertible syntaxin1A-mEos2 in the motor nerve terminal of Drosophila larvae by single-particle tracking photoactivation localization microscopy. Opto- and thermo-genetic neuronal stimulation increased syntaxin1A-mEos2 mobility, and reduced the size and molecular density of nanoclusters, suggesting an activity-dependent release of syntaxin1A from the confinement of nanoclusters. Syntaxin1A mobility was increased by mutating its polyphosphoinositide-binding site or preventing SNARE complex assembly via co-expression of tetanus toxin light chain. In contrast, syntaxin1A mobility was reduced by preventing SNARE complex disassembly. Our data demonstrate that polyphosphoinositide favours syntaxin1A trapping, and show that SNARE complex disassembly leads to syntaxin1A dissociation from nanoclusters. Lateral diffusion and trapping of syntaxin1A in nanoclusters therefore dynamically regulate neurotransmitter release. Syntaxin1A (Sx1A) is organized in nanoclusters in neurosecretory cells but how these nanoclusters are affected by neurotransmitter release in a living organism is unknown. Here the authors perform single molecule imaging analysis in live fly larvae and show that the lateral diffusion and trapping of Sx1A in nanoclusters are altered by synaptic activity.
Collapse
Affiliation(s)
- Adekunle T Bademosi
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elsa Lauwers
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, Leuven Institute for Neurodegenerative Disease (LIND), 3000 Leuven, Belgium
| | - Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lorenzo Odierna
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ye Jin Chai
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andreas Papadopulos
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Geoffrey J Goodhill
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.,School of Mathematics and Physics, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Patrik Verstreken
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, Leuven Institute for Neurodegenerative Disease (LIND), 3000 Leuven, Belgium
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
33
|
Chai YJ, Sierecki E, Tomatis VM, Gormal RS, Giles N, Morrow IC, Xia D, Götz J, Parton RG, Collins BM, Gambin Y, Meunier FA. Munc18-1 is a molecular chaperone for α-synuclein, controlling its self-replicating aggregation. J Cell Biol 2016; 214:705-18. [PMID: 27597756 PMCID: PMC5021092 DOI: 10.1083/jcb.201512016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 08/03/2016] [Indexed: 01/06/2023] Open
Abstract
Munc18-1 heterozygous mutations are associated with developmental diseases, including early infantile epileptic encephalopathy (EIEE). Chai et al. report that Munc18-1 acts as a chaperone for α-synuclein and controls its aggregative propensity. Munc18-1 EIEE-associated mutations promote the aggregation of endogenous α-synuclein in neurons, leading to a neurodegenerative phenotype. Munc18-1 is a key component of the exocytic machinery that controls neurotransmitter release. Munc18-1 heterozygous mutations cause developmental defects and epileptic phenotypes, including infantile epileptic encephalopathy (EIEE), suggestive of a gain of pathological function. Here, we used single-molecule analysis, gene-edited cells, and neurons to demonstrate that Munc18-1 EIEE-causing mutants form large polymers that coaggregate wild-type Munc18-1 in vitro and in cells. Surprisingly, Munc18-1 EIEE mutants also form Lewy body–like structures that contain α-synuclein (α-Syn). We reveal that Munc18-1 binds α-Syn, and its EIEE mutants coaggregate α-Syn. Likewise, removal of endogenous Munc18-1 increases the aggregative propensity of α-SynWT and that of the Parkinson’s disease–causing α-SynA30P mutant, an effect rescued by Munc18-1WT expression, indicative of chaperone activity. Coexpression of the α-SynA30P mutant with Munc18-1 reduced the number of α-SynA30P aggregates. Munc18-1 mutations and haploinsufficiency may therefore trigger a pathogenic gain of function through both the corruption of native Munc18-1 and a perturbed chaperone activity for α-Syn leading to aggregation-induced neurodegeneration.
Collapse
Affiliation(s)
- Ye Jin Chai
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Emma Sierecki
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia Single Molecule Sciences Centre, European Molecular Biology Laboratory Australia, The University of New South Wales, Sydney 2052, Australia
| | - Vanesa M Tomatis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nichole Giles
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia Single Molecule Sciences Centre, European Molecular Biology Laboratory Australia, The University of New South Wales, Sydney 2052, Australia
| | - Isabel C Morrow
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Di Xia
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yann Gambin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia Single Molecule Sciences Centre, European Molecular Biology Laboratory Australia, The University of New South Wales, Sydney 2052, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
34
|
Meunier FA, Gutiérrez LM. Captivating New Roles of F-Actin Cortex in Exocytosis and Bulk Endocytosis in Neurosecretory Cells. Trends Neurosci 2016; 39:605-613. [DOI: 10.1016/j.tins.2016.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/01/2016] [Accepted: 07/06/2016] [Indexed: 12/01/2022]
|
35
|
Gabel M, Chasserot-Golaz S. Annexin A2, an essential partner of the exocytotic process in chromaffin cells. J Neurochem 2016; 137:890-6. [DOI: 10.1111/jnc.13628] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/09/2016] [Accepted: 03/30/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Marion Gabel
- INCI; UPR3212 CNRS; Université de Strasbourg; Strasbourg France
| | | |
Collapse
|
36
|
Villanueva J, Gimenez-Molina Y, Viniegra S, Gutiérrez LM. F-actin cytoskeleton and the fate of organelles in chromaffin cells. J Neurochem 2016; 137:860-6. [PMID: 26843469 DOI: 10.1111/jnc.13560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/31/2022]
Abstract
In addition to playing a fundamental structural role, the F-actin cytoskeleton in neuroendocrine chromaffin cells has a prominent influence on governing the molecular mechanism and regulating the secretory process. Performing such roles, the F-actin network might be essential to first transport, and later locate the cellular organelles participating in the secretory cycle. Chromaffin granules are transported from the internal cytosolic regions to the cell periphery along microtubular and F-actin structures. Once in the cortical region, they are embedded in the F-actin network where these vesicles experience restrictions in motility. Similarly, mitochondria transport is affected by both microtubule and F-actin inhibitors and suffers increasing motion restrictions when they are located in the cortical region. Therefore, the F-actin cortex is a key factor in defining the existence of two populations of cortical and perinuclear granules and mitochondria which could be distinguished by their different location and mobility. Interestingly, other important organelles for controlling intracellular calcium levels, such as the endoplasmic reticulum network, present clear differences in distribution and much lower mobility than chromaffin vesicles and mitochondria. Nevertheless, both mitochondria and the endoplasmic reticulum appear to distribute in the proximity of secretory sites to fulfill a pivotal role, forming triads with calcium channels ensuring the fine tuning of the secretory response. This review presents the contributions that provide the basis for our current view regarding the influence that F-actin has on the distribution of organelles participating in the release of catecholamines in chromaffin cells, and summarizes this knowledge in simple models. In chromaffin cells, organelles such as granules and mitochondria distribute forming cortical and perinuclear populations whereas others like the ER present homogenous distributions. In the present review we discuss the role of transport systems and the existence of an F-actin cortical structure as the main factors behind the formation of organelle subpopulations in this neuroendocrine cell model. This article is part of a mini review series on Chromaffin cells (ISCCB Meeting, 2015). Cover image for this issue: doi: 10.1111/jnc.13322.
Collapse
Affiliation(s)
- José Villanueva
- Instituto de Neurociencias, Centro Mixto Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant (Alicante), Spain
| | - Yolanda Gimenez-Molina
- Instituto de Neurociencias, Centro Mixto Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant (Alicante), Spain
| | - Salvador Viniegra
- Instituto de Neurociencias, Centro Mixto Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant (Alicante), Spain
| | - Luis M Gutiérrez
- Instituto de Neurociencias, Centro Mixto Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant (Alicante), Spain
| |
Collapse
|
37
|
Heissler SM, Sellers JR. Kinetic Adaptations of Myosins for Their Diverse Cellular Functions. Traffic 2016; 17:839-59. [PMID: 26929436 DOI: 10.1111/tra.12388] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/18/2022]
Abstract
Members of the myosin superfamily are involved in all aspects of eukaryotic life. Their function ranges from the transport of organelles and cargos to the generation of membrane tension, and the contraction of muscle. The diversity of physiological functions is remarkable, given that all enzymatically active myosins follow a conserved mechanoenzymatic cycle in which the hydrolysis of ATP to ADP and inorganic phosphate is coupled to either actin-based transport or tethering of actin to defined cellular compartments. Kinetic capacities and limitations of a myosin are determined by the extent to which actin can accelerate the hydrolysis of ATP and the release of the hydrolysis products and are indispensably linked to its physiological tasks. This review focuses on kinetic competencies that - together with structural adaptations - result in myosins with unique mechanoenzymatic properties targeted to their diverse cellular functions.
Collapse
Affiliation(s)
- Sarah M Heissler
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, B50/3523, Bethesda, MD 20892-8015, USA
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, B50/3523, Bethesda, MD 20892-8015, USA
| |
Collapse
|
38
|
He F, Wollscheid HP, Nowicka U, Biancospino M, Valentini E, Ehlinger A, Acconcia F, Magistrati E, Polo S, Walters KJ. Myosin VI Contains a Compact Structural Motif that Binds to Ubiquitin Chains. Cell Rep 2016; 14:2683-94. [PMID: 26971995 DOI: 10.1016/j.celrep.2016.01.079] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/24/2015] [Accepted: 01/27/2016] [Indexed: 12/13/2022] Open
Abstract
Myosin VI is critical for cargo trafficking and sorting during early endocytosis and autophagosome maturation, and abnormalities in these processes are linked to cancers, neurodegeneration, deafness, and hypertropic cardiomyopathy. We identify a structured domain in myosin VI, myosin VI ubiquitin-binding domain (MyUb), that binds to ubiquitin chains, especially those linked via K63, K11, and K29. Herein, we solve the solution structure of MyUb and MyUb:K63-linked diubiquitin. MyUb folds as a compact helix-turn-helix-like motif and nestles between the ubiquitins of K63-linked diubiquitin, interacting with distinct surfaces of each. A nine-amino-acid extension at the C-terminal helix (Helix2) of MyUb is required for myosin VI interaction with endocytic and autophagic adaptors. Structure-guided mutations revealed that a functional MyUb is necessary for optineurin interaction. In addition, we found that an isoform-specific helix restricts MyUb binding to ubiquitin chains. This work provides fundamental insights into myosin VI interaction with ubiquitinated cargo and functional adaptors.
Collapse
Affiliation(s)
- Fahu He
- Protein Processing Section, Structural Biophysics Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Hans-Peter Wollscheid
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milano, Italy
| | - Urszula Nowicka
- Protein Processing Section, Structural Biophysics Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Matteo Biancospino
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milano, Italy
| | - Eleonora Valentini
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milano, Italy
| | - Aaron Ehlinger
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Filippo Acconcia
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milano, Italy
| | - Elisa Magistrati
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milano, Italy
| | - Simona Polo
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milano, Italy; DIPO, Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Via di Rudinì 8, 20122 Milan, Italy.
| | - Kylie J Walters
- Protein Processing Section, Structural Biophysics Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
39
|
Wollscheid HP, Biancospino M, He F, Magistrati E, Molteni E, Lupia M, Soffientini P, Rottner K, Cavallaro U, Pozzoli U, Mapelli M, Walters KJ, Polo S. Diverse functions of myosin VI elucidated by an isoform-specific α-helix domain. Nat Struct Mol Biol 2016; 23:300-308. [PMID: 26950368 PMCID: PMC4964928 DOI: 10.1038/nsmb.3187] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 02/09/2016] [Indexed: 01/01/2023]
Abstract
Myosin VI functions in endocytosis and cell motility. Alternative splicing of myosin VI mRNA generates two distinct isoform types, myosin VIshort and myosin VIlong, which differ in the C-terminal region. Their physiological and pathological role remains unknown. Here we identified an isoform-specific regulatory helix, named α2-linker that defines specific conformations and hence determines the target selectivity of human myosin VI. The presence of the α2-linker structurally defines a novel clathrin-binding domain that is unique to myosin VIlong and masks the known RRL interaction motif. This finding is relevant to ovarian cancer, where alternative myosin VI splicing is aberrantly regulated, and exon skipping dictates cell addiction to myosin VIshort for tumor cell migration. The RRL interactor optineurin contributes to this process by selectively binding myosin VIshort. Thus the α2-linker acts like a molecular switch that assigns myosin VI to distinct endocytic (myosin VIlong) or migratory (myosin VIshort) functional roles.
Collapse
Affiliation(s)
| | - Matteo Biancospino
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan 20139, Italy
| | - Fahu He
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Elisa Magistrati
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan 20139, Italy
| | - Erika Molteni
- Computational Biology, Scientific Institute IRCCS E.MEDEA, Bosisio Parini 23842, Italy
| | - Michela Lupia
- Molecular Medicine Program, European Institute of Oncology, Milan 20141, Italy
| | - Paolo Soffientini
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan 20139, Italy
| | - Klemens Rottner
- Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany.,Braunschweig University of Technology, 38106 Braunschweig, Germany
| | - Ugo Cavallaro
- Molecular Medicine Program, European Institute of Oncology, Milan 20141, Italy
| | - Uberto Pozzoli
- Computational Biology, Scientific Institute IRCCS E.MEDEA, Bosisio Parini 23842, Italy
| | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Kylie J Walters
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Simona Polo
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan 20139, Italy.,Dipartimento di oncologia ed emato-oncologia (DIPO), Universita' degli Studi di Milano, Milan 20122, Italy
| |
Collapse
|
40
|
|
41
|
Gabel M, Delavoie F, Demais V, Royer C, Bailly Y, Vitale N, Bader MF, Chasserot-Golaz S. Annexin A2-dependent actin bundling promotes secretory granule docking to the plasma membrane and exocytosis. J Cell Biol 2015; 210:785-800. [PMID: 26323692 PMCID: PMC4555831 DOI: 10.1083/jcb.201412030] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Annexin A2, a calcium-, actin-, and lipid-binding protein involved in exocytosis, mediates the formation of lipid microdomains required for the structural and spatial organization of fusion sites at the plasma membrane. To understand how annexin A2 promotes this membrane remodeling, the involvement of cortical actin filaments in lipid domain organization was investigated. 3D electron tomography showed that cortical actin bundled by annexin A2 connected docked secretory granules to the plasma membrane and contributed to the formation of GM1-enriched lipid microdomains at the exocytotic sites in chromaffin cells. When an annexin A2 mutant with impaired actin filament-bundling activity was expressed, the formation of plasma membrane lipid microdomains and the number of exocytotic events were decreased and the fusion kinetics were slower, whereas the pharmacological activation of the intrinsic actin-bundling activity of endogenous annexin A2 had the opposite effects. Thus, annexin A2-induced actin bundling is apparently essential for generating active exocytotic sites.
Collapse
Affiliation(s)
- Marion Gabel
- Institut des Neurosciences Cellulaires et Intégratives, UPR3212 Centre National de la Recherche Scientifique, Université de Strasbourg, F-67084 Strasbourg, France
| | - Franck Delavoie
- Laboratoire de Biologie Moléculaire Eucaryote, UMR5099 Centre National de la Recherche Scientifique-Université de Toulouse III Paul Sabatier, F-31000 Toulouse, France
| | - Valérie Demais
- Plateforme Imagerie In Vitro, Neuropôle de Strasbourg, F-67084 Strasbourg, France
| | - Cathy Royer
- Plateforme Imagerie In Vitro, Neuropôle de Strasbourg, F-67084 Strasbourg, France
| | - Yannick Bailly
- Institut des Neurosciences Cellulaires et Intégratives, UPR3212 Centre National de la Recherche Scientifique, Université de Strasbourg, F-67084 Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, UPR3212 Centre National de la Recherche Scientifique, Université de Strasbourg, F-67084 Strasbourg, France
| | - Marie-France Bader
- Institut des Neurosciences Cellulaires et Intégratives, UPR3212 Centre National de la Recherche Scientifique, Université de Strasbourg, F-67084 Strasbourg, France
| | - Sylvette Chasserot-Golaz
- Institut des Neurosciences Cellulaires et Intégratives, UPR3212 Centre National de la Recherche Scientifique, Université de Strasbourg, F-67084 Strasbourg, France
| |
Collapse
|
42
|
Narayana V, Tomatis VM, Wang T, Kvaskoff D, Meunier F. Profiling of Free Fatty Acids Using Stable Isotope Tagging Uncovers a Role for Saturated Fatty Acids in Neuroexocytosis. ACTA ACUST UNITED AC 2015; 22:1552-1561. [DOI: 10.1016/j.chembiol.2015.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/02/2015] [Accepted: 09/24/2015] [Indexed: 01/18/2023]
|
43
|
Sundaramoorthy V, Walker AK, Tan V, Fifita JA, Mccann EP, Williams KL, Blair IP, Guillemin GJ, Farg MA, Atkin JD. Defects in optineurin- and myosin VI-mediated cellular trafficking in amyotrophic lateral sclerosis. Hum Mol Genet 2015; 24:3830-46. [DOI: 10.1093/hmg/ddv126] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022] Open
|
44
|
Papadopulos A, Gomez GA, Martin S, Jackson J, Gormal RS, Keating DJ, Yap AS, Meunier FA. Activity-driven relaxation of the cortical actomyosin II network synchronizes Munc18-1-dependent neurosecretory vesicle docking. Nat Commun 2015; 6:6297. [DOI: 10.1038/ncomms7297] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 01/14/2015] [Indexed: 01/08/2023] Open
|
45
|
Martin S, Papadopulos A, Tomatis VM, Sierecki E, Malintan NT, Gormal RS, Giles N, Johnston WA, Alexandrov K, Gambin Y, Collins BM, Meunier FA. Increased polyubiquitination and proteasomal degradation of a Munc18-1 disease-linked mutant causes temperature-sensitive defect in exocytosis. Cell Rep 2014; 9:206-218. [PMID: 25284778 DOI: 10.1016/j.celrep.2014.08.059] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 07/31/2014] [Accepted: 08/23/2014] [Indexed: 12/23/2022] Open
Abstract
Munc18-1 is a critical component of the core machinery controlling neuroexocytosis. Recently, mutations in Munc18-1 leading to the development of early infantile epileptic encephalopathy have been discovered. However, which degradative pathway controls Munc18-1 levels and how it impacts on neuroexocytosis in this pathology is unknown. Using neurosecretory cells deficient in Munc18, we show that a disease-linked mutation, C180Y, renders the protein unstable at 37°C. Although the mutated protein retains its function as t-SNARE chaperone, neuroexocytosis is impaired, a defect that can be rescued at a lower permissive temperature. We reveal that Munc18-1 undergoes K48-linked polyubiquitination, which is highly increased by the mutation, leading to proteasomal, but not lysosomal, degradation. Our data demonstrate that functional Munc18-1 levels are controlled through polyubiquitination and proteasomal degradation. The C180Y disease-causing mutation greatly potentiates this degradative pathway, rendering Munc18-1 unable to facilitate neuroexocytosis, a phenotype that is reversed at a permissive temperature.
Collapse
Affiliation(s)
- Sally Martin
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Andreas Papadopulos
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Vanesa M Tomatis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Emma Sierecki
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nancy T Malintan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nichole Giles
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Wayne A Johnston
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kirill Alexandrov
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yann Gambin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frederic A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
46
|
Wolny M, Batchelor M, Knight PJ, Paci E, Dougan L, Peckham M. Stable single α-helices are constant force springs in proteins. J Biol Chem 2014; 289:27825-35. [PMID: 25122759 PMCID: PMC4183817 DOI: 10.1074/jbc.m114.585679] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Single α-helix (SAH) domains are rich in charged residues (Arg, Lys, and Glu) and stable in solution over a wide range of pH and salt concentrations. They are found in many different proteins where they bridge two functional domains. To test the idea that their high stability might enable these proteins to resist unfolding along their length, the properties and unfolding behavior of the predicted SAH domain from myosin-10 were characterized. The expressed and purified SAH domain was highly helical, melted non-cooperatively, and was monomeric as shown by circular dichroism and mass spectrometry as expected for a SAH domain. Single molecule force spectroscopy experiments showed that the SAH domain unfolded at very low forces (<30 pN) without a characteristic unfolding peak. Molecular dynamics simulations showed that the SAH domain unfolds progressively as the length is increased and refolds progressively as the length is reduced. This enables the SAH domain to act as a constant force spring in the mechanically dynamic environment of the cell.
Collapse
Affiliation(s)
- Marcin Wolny
- From the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Matthew Batchelor
- From the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Peter J Knight
- From the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Emanuele Paci
- From the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Lorna Dougan
- From the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Michelle Peckham
- From the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
47
|
Olivares MJ, González-Jamett AM, Guerra MJ, Baez-Matus X, Haro-Acuña V, Martínez-Quiles N, Cárdenas AM. Src kinases regulate de novo actin polymerization during exocytosis in neuroendocrine chromaffin cells. PLoS One 2014; 9:e99001. [PMID: 24901433 PMCID: PMC4047038 DOI: 10.1371/journal.pone.0099001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 05/09/2014] [Indexed: 11/19/2022] Open
Abstract
The cortical actin network is dynamically rearranged during secretory processes. Nevertheless, it is unclear how de novo actin polymerization and the disruption of the preexisting actin network control transmitter release. Here we show that in bovine adrenal chromaffin cells, both formation of new actin filaments and disruption of the preexisting cortical actin network are induced by Ca2+ concentrations that trigger exocytosis. These two processes appear to regulate different stages of exocytosis; whereas the inhibition of actin polymerization with the N-WASP inhibitor wiskostatin restricts fusion pore expansion, thus limiting the release of transmitters, the disruption of the cortical actin network with cytochalasin D increases the amount of transmitter released per event. Further, the Src kinase inhibitor PP2, and cSrc SH2 and SH3 domains also suppress Ca2+-dependent actin polymerization, and slow down fusion pore expansion without disturbing the cortical F-actin organization. Finally, the isolated SH3 domain of c-Src prevents both the disruption of the actin network and the increase in the quantal release induced by cytochalasin D. These findings support a model where a rise in the cytosolic Ca2+ triggers actin polymerization through a mechanism that involves Src kinases. The newly formed actin filaments would speed up the expansion of the initial fusion pore, whereas the preexisting actin network might control a different step of the exocytosis process.
Collapse
Affiliation(s)
- María José Olivares
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Playa Ancha, Valparaíso, Chile
| | - Arlek M. González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Playa Ancha, Valparaíso, Chile
| | - María José Guerra
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Playa Ancha, Valparaíso, Chile
| | - Ximena Baez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Playa Ancha, Valparaíso, Chile
| | - Valentina Haro-Acuña
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Playa Ancha, Valparaíso, Chile
| | - Narcisa Martínez-Quiles
- Departamento de Microbiología (Inmunología), Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana M. Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Playa Ancha, Valparaíso, Chile
- * E-mail:
| |
Collapse
|
48
|
Low PC, Manzanero S, Mohannak N, Narayana VK, Nguyen TH, Kvaskoff D, Brennan FH, Ruitenberg MJ, Gelderblom M, Magnus T, Kim HA, Broughton BRS, Sobey CG, Vanhaesebroeck B, Stow JL, Arumugam TV, Meunier FA. PI3Kδ inhibition reduces TNF secretion and neuroinflammation in a mouse cerebral stroke model. Nat Commun 2014; 5:3450. [PMID: 24625684 DOI: 10.1038/ncomms4450] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/13/2014] [Indexed: 01/01/2023] Open
Abstract
Stroke is a major cause of death worldwide and the leading cause of permanent disability. Although reperfusion is currently used as treatment, the restoration of blood flow following ischaemia elicits a profound inflammatory response mediated by proinflammatory cytokines such as tumour necrosis factor (TNF), exacerbating tissue damage and worsening the outcomes for stroke patients. Phosphoinositide 3-kinase delta (PI3Kδ) controls intracellular TNF trafficking in macrophages and therefore represents a prospective target to limit neuroinflammation. Here we show that PI3Kδ inhibition confers protection in ischaemia/reperfusion models of stroke. In vitro, restoration of glucose supply following an episode of glucose deprivation potentiates TNF secretion from primary microglia-an effect that is sensitive to PI3Kδ inhibition. In vivo, transient middle cerebral artery occlusion and reperfusion in kinase-dead PI3Kδ (p110δ(D910A/D910A)) or wild-type mice pre- or post-treated with the PI3Kδ inhibitor CAL-101, leads to reduced TNF levels, decreased leukocyte infiltration, reduced infarct size and improved functional outcome. These data identify PI3Kδ as a potential therapeutic target in ischaemic stroke.
Collapse
Affiliation(s)
- Pei Ching Low
- 1] Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia [2]
| | - Silvia Manzanero
- 1] School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia [2]
| | - Nika Mohannak
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vinod K Narayana
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Tam H Nguyen
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David Kvaskoff
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Faith H Brennan
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Marc J Ruitenberg
- 1] Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia [2] School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hyun Ah Kim
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Brad R S Broughton
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Christopher G Sobey
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London WC1E 6DD, UK
| | - Jennifer L Stow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Thiruma V Arumugam
- 1] School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia [2]
| | - Frédéric A Meunier
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
49
|
Secretagogue stimulation of neurosecretory cells elicits filopodial extensions uncovering new functional release sites. J Neurosci 2014; 33:19143-53. [PMID: 24305811 DOI: 10.1523/jneurosci.2634-13.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Regulated exocytosis in neurosecretory cells relies on the timely fusion of secretory granules (SGs) with the plasma membrane. Secretagogue stimulation leads to an enlargement of the cell footprint (surface area in contact with the coverslip), an effect previously attributed to exocytic fusion of SGs with the plasma membrane. Using total internal reflection fluorescence microscopy, we reveal the formation of filopodia-like structures in bovine chromaffin and PC12 cells driving the footprint expansion, suggesting the involvement of cortical actin network remodeling in this process. Using exocytosis-incompetent PC12 cells, we demonstrate that footprint enlargement is largely independent of SG fusion, suggesting that vesicular exocytic fusion plays a relatively minor role in filopodial expansion. The footprint periphery, including filopodia, undergoes extensive F-actin remodeling, an effect abolished by the actomyosin inhibitors cytochalasin D and blebbistatin. Imaging of both Lifeact-GFP and the SG marker protein neuropeptide Y-mCherry reveals that SGs actively translocate along newly forming actin tracks before undergoing fusion. Together, these data demonstrate that neurosecretory cells regulate the number of SGs undergoing exocytosis during sustained stimulation by controlling vesicular mobilization and translocation to the plasma membrane through actin remodeling. Such remodeling facilitates the de novo formation of fusion sites.
Collapse
|
50
|
Maucort G, Kasula R, Papadopulos A, Nieminen TA, Rubinsztein-Dunlop H, Meunier FA. Mapping organelle motion reveals a vesicular conveyor belt spatially replenishing secretory vesicles in stimulated chromaffin cells. PLoS One 2014; 9:e87242. [PMID: 24489879 PMCID: PMC3906151 DOI: 10.1371/journal.pone.0087242] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/19/2013] [Indexed: 12/18/2022] Open
Abstract
How neurosecretory cells spatially adjust their secretory vesicle pools to replenish those that have fused and released their hormonal content is currently unknown. Here we designed a novel set of image analyses to map the probability of tracked organelles undergoing a specific type of movement (free, caged or directed). We then applied our analysis to time-lapse z-stack confocal imaging of secretory vesicles from bovine Chromaffin cells to map the global changes in vesicle motion and directionality occurring upon secretagogue stimulation. We report a defined region abutting the cortical actin network that actively transports secretory vesicles and is dissipated by actin and microtubule depolymerizing drugs. The directionality of this “conveyor belt” towards the cell surface is activated by stimulation. Actin and microtubule networks therefore cooperatively probe the microenvironment to transport secretory vesicles to the periphery, providing a mechanism whereby cells globally adjust their vesicle pools in response to secretagogue stimulation.
Collapse
Affiliation(s)
- Guillaume Maucort
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
- School of Mathematics and Physics, The University of Queensland, Brisbane, Queensland, Australia
| | - Ravikiran Kasula
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Andreas Papadopulos
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Timo A. Nieminen
- School of Mathematics and Physics, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Frederic A. Meunier
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|