51
|
Lin CC, Hsu HF, Walla PJ. A One Donor-Two Acceptor Lipid Bilayer FRET Assay Based on Asymmetrically Labeled Liposomes. J Phys Chem B 2016; 120:11085-11092. [PMID: 27762543 DOI: 10.1021/acs.jpcb.6b05654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The fusion of two opposing membranes is essential in biological functions such as fertilization, viral entry, membrane trafficking and synaptic transmission. Before the membrane bilayers are fully connected, at some stage a hemifusion intermediate-when the outer leaflets are merged but not the inner leaflets-is formed. However, the position of hemifusion in the energy landscape and the duration of it vary and have not been fully mapped out. To date, there has not been a way to differentiate lipid mixing of the two leaflets directly in a single experiment. Herein we demonstrate labeling of the outer and inner leaflets with different fluorophores, which can be distinguished by their fluorescence lifetimes. As a proof of concept, the asymmetrically labeled liposomes were used as acceptor liposomes in a novel one donor-two acceptor Förster resonance energy transfer (FRET) assay to monitor membrane fusion reactions mediated by the synaptic proteins soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) in microfluidic devices. Initial hemifusion was clearly indicated by the acceptor fluorescence lifetime originating solely from FRET acceptors on the outer leaflet (Oregon Green 488; τFl ∼ 4.8 ns). Progression to full fusion was then indicated by the significantly increasing lifetime contribution from acceptors on the inner leaflet (nitrobenzoxadiazole; τFl ∼ 6.7 ns). The new labeling strategy creates many possibilities in the design of bulk and single-molecule experiments.
Collapse
Affiliation(s)
- Chao-Chen Lin
- Research Group Biomolecular Spectroscopy and Single-Molecule Detection, Max Planck Institute for Biophysical Chemistry , Am Faßberg 11, 37077 Göttingen, Germany
| | - Hsin-Fang Hsu
- Laboratory for Fluid Dynamics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization , Am Faßberg 17, 37077 Göttingen, Germany
| | - Peter Jomo Walla
- Research Group Biomolecular Spectroscopy and Single-Molecule Detection, Max Planck Institute for Biophysical Chemistry , Am Faßberg 11, 37077 Göttingen, Germany.,Department of Biophysical Chemistry, Institute for Physical and Theoretical Chemistry, Technical University of Braunschweig , Hans-Sommer-Straße 10, 38106 Braunschweig, Germany
| |
Collapse
|
52
|
Lee H, Jin W, Jeong BC, Suh JW. A new in vitro hemagglutinin inhibitor screening system based on a single-vesicle fusion assay. Sci Rep 2016; 6:30642. [PMID: 27469068 PMCID: PMC4965830 DOI: 10.1038/srep30642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/07/2016] [Indexed: 11/09/2022] Open
Abstract
Hemagglutinin (HA) from the influenza virus plays a pivotal role in the infection of host mammalian cells and is, therefore, a druggable target, similar to neuraminidase. However, research involving the influenza virus must be conducted in facilities certified at or above Biosafety Level 2 because of the potential threat of the contagiousness of this virus. To develop a new HA inhibitor screening system without intact influenza virus, we conceived a single-vesicle fusion assay using full-length recombinant HA. In this study, we first showed that full-length recombinant HA can mediate membrane fusion in ensemble and single-vesicle fusion assays. The fluorescence resonance energy transfer (FRET) frequency pattern of single-vesicle complexes completely differed when the inhibitors targeted the HA1 or HA2 domain of HA. This result indicates that analysing the FRET patterns in this assay can provide information regarding the domains of HA inhibited by compounds and compounds' inhibitory activities. Therefore, our results suggest that the assay developed here is a promising tool for the discovery of anti-influenza virus drug candidates as a new in vitro inhibitor screening system against HA from the influenza virus.
Collapse
Affiliation(s)
- Hanki Lee
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Yongin, Gyeonggi-do, 17058, Republic of Korea
| | - Wook Jin
- Laboratory of Molecular Disease and Cell Regulation, Department of Molecular Medicine, School of Medicine, Gacheon University, Incheon, 21936, Republic of Korea.,Gachon Medical Research Institute, Gil Medical Center, Incheon, 21565, Republic of Korea
| | - Byeong-Chul Jeong
- Division of Biosciences and Bioinformatics, College of Natural Science, Myongji University, Yongin, Gyeonggi-do, 17058, Republic of Korea
| | - Joo-Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Yongin, Gyeonggi-do, 17058, Republic of Korea.,Division of Biosciences and Bioinformatics, College of Natural Science, Myongji University, Yongin, Gyeonggi-do, 17058, Republic of Korea
| |
Collapse
|
53
|
Kim J, Zhu Y, Shin YK. Preincubation of t-SNAREs with Complexin I Increases Content-Mixing Efficiency. Biochemistry 2016; 55:3667-73. [PMID: 27286417 DOI: 10.1021/acs.biochem.6b00114] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Complexin (Cpx) is a major regulator for Ca(2+)-triggered fast neuroexocytosis which underlies neuronal communication. Many psychiatric and neurological disorders accompany changes in the Cpx expression level, suggesting that abnormal Cpx levels may elicit aberrant cognitive symptoms. To comprehend how the changes in the Cpx level might affect neuronal communication, we investigated Ca(2+)-triggered exocytosis at various Cpx concentrations. Ca(2+)-triggered content-mixing between a single proteoliposome of t-SNARE and another single proteoliposome of v-SNARE plus Ca(2+)-sensor synaptotagmin 1 was examined with total internal reflection microscopy. We find that Cpx enhances Ca(2+)-triggered vesicle fusion with the yield changing from approximately 10% to 70% upon increasing Cpx from 0 to 100 nM. Unexpectedly, however, the fusion efficiency becomes reduced when Cpx is increased further, dropping to 20% in the micromolar range, revealing a bell-shaped dose-response curve. Intriguingly, we find that the rate of vesicle fusion is nearly invariant through the entire range of Cpx concentrations studied, suggesting that a reevaluation of the current Cpx clamping mechanism is necessary. Thus, our results provide insights into how delicately Cpx fine-tunes neuronal communication.
Collapse
Affiliation(s)
- Jaewook Kim
- Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University , Ames, Iowa 50011, United States
| | - Yicheng Zhu
- Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University , Ames, Iowa 50011, United States
| | - Yeon-Kyun Shin
- Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University , Ames, Iowa 50011, United States
| |
Collapse
|
54
|
Nanodisc-cell fusion: control of fusion pore nucleation and lifetimes by SNARE protein transmembrane domains. Sci Rep 2016; 6:27287. [PMID: 27264104 PMCID: PMC4893671 DOI: 10.1038/srep27287] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/13/2016] [Indexed: 12/26/2022] Open
Abstract
The initial, nanometer-sized connection between the plasma membrane and a hormone- or neurotransmitter-filled vesicle –the fusion pore– can flicker open and closed repeatedly before dilating or resealing irreversibly. Pore dynamics determine release and vesicle recycling kinetics, but pore properties are poorly known because biochemically defined single-pore assays are lacking. We isolated single flickering pores connecting v-SNARE-reconstituted nanodiscs to cells ectopically expressing cognate, “flipped” t-SNAREs. Conductance through single, voltage-clamped fusion pores directly reported sub-millisecond pore dynamics. Pore currents fluctuated, transiently returned to baseline multiple times, and disappeared ~6 s after initial opening, as if the fusion pore fluctuated in size, flickered, and resealed. We found that interactions between v- and t-SNARE transmembrane domains (TMDs) promote, but are not essential for pore nucleation. Surprisingly, TMD modifications designed to disrupt v- and t-SNARE TMD zippering prolonged pore lifetimes dramatically. We propose that the post-fusion geometry of the proteins contribute to pore stability.
Collapse
|
55
|
Heo P, Yang Y, Han KY, Kong B, Shin JH, Jung Y, Jeong C, Shin J, Shin YK, Ha T, Kweon DH. A Chemical Controller of SNARE-Driven Membrane Fusion That Primes Vesicles for Ca(2+)-Triggered Millisecond Exocytosis. J Am Chem Soc 2016; 138:4512-21. [PMID: 26987363 PMCID: PMC4852477 DOI: 10.1021/jacs.5b13449] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Membrane fusion is mediated by the SNARE complex which is formed through a zippering process. Here, we developed a chemical controller for the progress of membrane fusion. A hemifusion state was arrested by a polyphenol myricetin which binds to the SNARE complex. The arrest of membrane fusion was rescued by an enzyme laccase that removes myricetin from the SNARE complex. The rescued hemifusion state was metastable and long-lived with a decay constant of 39 min. This membrane fusion controller was applied to delineate how Ca(2+) stimulates fusion-pore formation in a millisecond time scale. We found, using a single-vesicle fusion assay, that such myricetin-primed vesicles with synaptotagmin 1 respond synchronously to physiological concentrations of Ca(2+). When 10 μM Ca(2+) was added to the hemifused vesicles, the majority of vesicles rapidly advanced to fusion pores with a time constant of 16.2 ms. Thus, the results demonstrate that a minimal exocytotic membrane fusion machinery composed of SNAREs and synaptotagmin 1 is capable of driving membrane fusion in a millisecond time scale when a proper vesicle priming is established. The chemical controller of SNARE-driven membrane fusion should serve as a versatile tool for investigating the differential roles of various synaptic proteins in discrete fusion steps.
Collapse
Affiliation(s)
- Paul Heo
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, and Center for Human Interface Nano Technology, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, South Korea
| | - Yoosoo Yang
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, and Center for Human Interface Nano Technology, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, South Korea
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - Kyu-Young Han
- Howard Hughes Medical Institute, Baltimore, MD 21205, United States
- CREOL, The College of Optics & Photonics, University of Central Florida, Orlando, Florida 32816, United States
| | - Byoungjae Kong
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, and Center for Human Interface Nano Technology, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, South Korea
| | - Jong-Hyeok Shin
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, and Center for Human Interface Nano Technology, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, South Korea
| | - Younghoon Jung
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, and Center for Human Interface Nano Technology, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, South Korea
| | - Cherlhyun Jeong
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - Jaeil Shin
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, United States
| | - Yeon-Kyun Shin
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, United States
| | - Taekjip Ha
- Howard Hughes Medical Institute, Baltimore, MD 21205, United States
- Department of Biophysics and Biophysical Chemistry Johns Hopkins University School of Medicine, Baltimore, MD 21205 United States
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218 United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Dae-Hyuk Kweon
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, and Center for Human Interface Nano Technology, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, South Korea
| |
Collapse
|
56
|
Xu W, Nathwani B, Lin C, Wang J, Karatekin E, Pincet F, Shih W, Rothman JE. A Programmable DNA Origami Platform to Organize SNAREs for Membrane Fusion. J Am Chem Soc 2016; 138:4439-47. [PMID: 26938705 DOI: 10.1021/jacs.5b13107] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes are the core molecular machinery of membrane fusion, a fundamental process that drives inter- and intracellular communication and trafficking. One of the questions that remains controversial has been whether and how SNAREs cooperate. Here we show the use of self-assembled DNA-nanostructure rings to template uniform-sized small unilamellar vesicles containing predetermined maximal number of externally facing SNAREs to study the membrane-fusion process. We also incorporated lipid-conjugated complementary ssDNA as tethers into vesicle and target membranes, which enabled bypass of the rate-limiting docking step of fusion reactions and allowed direct observation of individual membrane-fusion events at SNARE densities as low as one pair per vesicle. With this platform, we confirmed at the single event level that, after docking of the templated-SUVs to supported lipid bilayers (SBL), one to two pairs of SNAREs are sufficient to drive fast lipid mixing. Modularity and programmability of this platform makes it readily amenable to studying more complicated systems where auxiliary proteins are involved.
Collapse
Affiliation(s)
| | - Bhavik Nathwani
- Wyss Institute for Biologically Inspired Engineering and Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Department of Cancer Biology, Dana Farber Cancer Institute , Boston, Massachusetts 02115, United States
| | - Chenxiang Lin
- Wyss Institute for Biologically Inspired Engineering and Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Department of Cancer Biology, Dana Farber Cancer Institute , Boston, Massachusetts 02115, United States
| | | | - Erdem Karatekin
- Laboratoire de Neurophotonique, Université Paris Descartes, Faculté des Sciences Fondamentales et Biomédicales, Centre National de la Recherche Scientifique (CNRS) UMR8250, 45, rue des Saints Pères, 75270 Cedex 06 Paris, France
| | - Frederic Pincet
- Laboratoire de Physique Statistique, Ecole Normale Supérieure de Paris, Université Pierre et Marie Curie, Université Paris Diderot, Centre National de la Recherche Scientifique, UMR 8550, 24 rue Lhomond, 75005 Paris, France
| | - William Shih
- Wyss Institute for Biologically Inspired Engineering and Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Department of Cancer Biology, Dana Farber Cancer Institute , Boston, Massachusetts 02115, United States
| | | |
Collapse
|
57
|
Shin J, Jung YH, Cho DH, Park M, Lee KE, Yang Y, Jeong C, Sung BH, Sohn JH, Park JB, Kweon DH. Display of membrane proteins on the heterologous caveolae carved by caveolin-1 in the Escherichia coli cytoplasm. Enzyme Microb Technol 2015; 79-80:55-62. [DOI: 10.1016/j.enzmictec.2015.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/20/2015] [Accepted: 06/26/2015] [Indexed: 11/25/2022]
|
58
|
Mahmoodzadeh Hosseini H, Halabian R, Amin M, Imani Fooladi AA. Texosome-based drug delivery system for cancer therapy: from past to present. Cancer Biol Med 2015; 12:150-62. [PMID: 26487960 PMCID: PMC4607826 DOI: 10.7497/j.issn.2095-3941.2015.0045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rising worldwide cancer incidence and resistance to current anti-cancer drugs necessitate the need for new pharmaceutical compounds and drug delivery system. Malfunction of the immune system, particularly in the tumor microenvironment, causes tumor growth and enhances tumor progression. Thus, cancer immunotherapy can be an appropriate approach to provoke the systemic immune system to combat tumor expansion. Texosomes, which are endogenous nanovesicles released by all tumor cells, contribute to cell-cell communication and modify the phenotypic features of recipient cells due to the texosomes' ability to transport biological components. For this reason, texosome-based delivery system can be a valuable strategy for therapeutic purposes. To improve the pharmaceutical behavior of this system and to facilitate its use in medical applications, biotechnology approaches and mimetic techniques have been utilized. In this review, we present the development history of texosome-based delivery systems and discuss the advantages and disadvantages of each system.
Collapse
Affiliation(s)
- Hamideh Mahmoodzadeh Hosseini
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| | - Raheleh Halabian
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| | - Mohsen Amin
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| | - Abbas Ali Imani Fooladi
- 1 Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran ; 2 Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653861, Iran
| |
Collapse
|
59
|
Yang Y, Kim J, Kim HY, Ryoo N, Lee S, Kim Y, Rhim H, Shin YK. Amyloid-β Oligomers May Impair SNARE-Mediated Exocytosis by Direct Binding to Syntaxin 1a. Cell Rep 2015; 12:1244-51. [PMID: 26279571 DOI: 10.1016/j.celrep.2015.07.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 06/17/2015] [Accepted: 07/22/2015] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is closely associated with synaptic dysfunction, and thus current treatments often aim to stimulate neurotransmission to improve cognitive impairment. Whereas the formation of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is essential for synaptic transmission, the correlation between SNAREs and AD neuropathology is unknown. Here, we report that intracellular amyloid-β (Aβ) oligomers directly inhibit SNARE-mediated exocytosis by impairing SNARE complex formation. We observe abnormal reduction of SNARE complex levels in the brains of APP/PS1 transgenic (TG) mice compared to age-matched wild-types. We demonstrate that Aβ oligomers block SNARE complex assembly through the direct interaction with a target membrane (t)-SNARE syntaxin 1a in vitro. Furthermore, the results of the in vitro single-vesicle content-mixing assay reveal that Aβ oligomers inhibit SNARE-mediated fusion pores. Thus, our study identifies a potential molecular mechanism by which intracellular Aβ oligomers hamper SNARE-mediated exocytosis, likely leading to AD-associated synaptic dysfunctions.
Collapse
Affiliation(s)
- Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Jaewook Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea; Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Hye Yun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Nayeon Ryoo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Sejin Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - YoungSoo Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Hyewhon Rhim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea; Department of Neuroscience, University of Science and Technology (UST), Daejon 305-333, Republic of Korea.
| | - Yeon-Kyun Shin
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea; Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
60
|
Gong B, Choi BK, Kim JY, Shetty D, Ko YH, Selvapalam N, Lee NK, Kim K. High Affinity Host-Guest FRET Pair for Single-Vesicle Content-Mixing Assay: Observation of Flickering Fusion Events. J Am Chem Soc 2015; 137:8908-11. [PMID: 26160008 DOI: 10.1021/jacs.5b05385] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fluorescence-based single-vesicle fusion assays provide a powerful method for studying mechanisms underlying complex biological processes of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)-mediated vesicle fusion and neurotransmitter release. A crucial element of these assays is the ability of the fluorescent probe(s) to reliably detect key intermediate events of fusion pore opening and content release/mixing. Here, we report a new, reliable, and efficient single-vesicle content-mixing assay using a high affinity, fluorophore tagged host-guest pair, cucurbit[7]uril-Cy3 and adamantane-Cy5 as a fluorescence resonance energy transfer (FRET) pair. The power of these probes is demonstrated by the first successful observation of flickering dynamics of the fusion pore by in vitro assay using neuronal SNARE-reconstituted vesicles.
Collapse
Affiliation(s)
- Bokyoung Gong
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Bong-Kyu Choi
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Jae-Yeol Kim
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Dinesh Shetty
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Young Ho Ko
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Narayanan Selvapalam
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Nam Ki Lee
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Kimoon Kim
- †Center for Self-Assembly and Complexity (CSC), Institute for Basic Science (IBS), ‡Department of Chemistry, §School of Interdisciplinary Bioscience and Bioengineering, and ∥Department of Physics, #Division of Advanced Materials Science, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| |
Collapse
|
61
|
Schwenen LLG, Hubrich R, Milovanovic D, Geil B, Yang J, Kros A, Jahn R, Steinem C. Resolving single membrane fusion events on planar pore-spanning membranes. Sci Rep 2015; 5:12006. [PMID: 26165860 PMCID: PMC4499801 DOI: 10.1038/srep12006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/15/2015] [Indexed: 12/15/2022] Open
Abstract
Even though a number of different in vitro fusion assays have been developed to analyze protein mediated fusion, they still only partially capture the essential features of the in vivo situation. Here we established an in vitro fusion assay that mimics the fluidity and planar geometry of the cellular plasma membrane to be able to monitor fusion of single protein-containing vesicles. As a proof of concept, planar pore-spanning membranes harboring SNARE-proteins were generated on highly ordered functionalized 1.2 μm-sized pore arrays in Si3N4. Full mobility of the membrane components was demonstrated by fluorescence correlation spectroscopy. Fusion was analyzed by two color confocal laser scanning fluorescence microscopy in a time resolved manner allowing to readily distinguish between vesicle docking, intermediate states such as hemifusion and full fusion. The importance of the membrane geometry on the fusion process was highlighted by comparing SNARE-mediated fusion with that of a minimal SNARE fusion mimetic.
Collapse
Affiliation(s)
- Lando L G Schwenen
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany
| | - Raphael Hubrich
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany
| | - Dragomir Milovanovic
- Max-Planck-Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Burkhard Geil
- Institute for Physical Chemistry, University of Göttingen, Tammannstr. 6, 37077 Göttingen, Germany
| | - Jian Yang
- Leiden Institute of Chemistry - Supramolecular and Biomaterials Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Alexander Kros
- Leiden Institute of Chemistry - Supramolecular and Biomaterials Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Reinhard Jahn
- Max-Planck-Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Claudia Steinem
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Tammannstr. 2, 37077 Göttingen, Germany
| |
Collapse
|
62
|
Ishitsuka Y, Azadfar N, Kobitski AY, Nienhaus K, Johnsson N, Nienhaus GU. Evaluation of Genetically Encoded Chemical Tags as Orthogonal Fluorophore Labeling Tools for Single-Molecule FRET Applications. J Phys Chem B 2015; 119:6611-9. [PMID: 25978145 DOI: 10.1021/acs.jpcb.5b03584] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fluorescence resonance energy transfer (FRET) is a superb technique for measuring conformational changes of proteins on the single molecule level (smFRET) in real time. It requires introducing a donor and acceptor fluorophore pair at specific locations on the protein molecule of interest, which has often been a challenging task. By using two different self-labeling chemical tags, such as Halo-, TMP-, SNAP- and CLIP-tags, orthogonal labeling may be achieved rapidly and reliably. However, these comparatively large tags add extra distance and flexibility between the desired labeling location on the protein and the fluorophore position, which may affect the results. To systematically characterize chemical tags for smFRET measurement applications, we took the SNAP-tag/CLIP-tag combination as a model system and fused a flexible unstructured peptide, rigid polyproline peptides of various lengths, and the calcium sensor protein calmodulin between the tags. We could reliably identify length variations as small as four residues in the polyproline peptide. In the calmodulin system, the added length introduced by these tags was even beneficial for revealing subtle conformational changes upon variation of the buffer conditions. This approach opens up new possibilities for studying conformational dynamics, especially in large protein systems that are difficult to specifically conjugate with fluorophores.
Collapse
Affiliation(s)
- Yuji Ishitsuka
- †Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Naghmeh Azadfar
- †Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Andrei Yu Kobitski
- †Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Karin Nienhaus
- †Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Strasse 1, 76131 Karlsruhe, Germany
| | - Nils Johnsson
- §Institute of Molecular Genetics and Cell Biology, Ulm University, James Franck Ring N27, 89081 Ulm, Germany
| | - G Ulrich Nienhaus
- †Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Strasse 1, 76131 Karlsruhe, Germany.,∥Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.,⊥Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, Illinois 61801, United States
| |
Collapse
|
63
|
Lai Y, Zhao L, Bu B, Lou X, Li D, Ji B, Liu J, Diao J, Shin YK. Lipid molecules influence early stages of yeast SNARE-mediated membrane fusion. Phys Biol 2015; 12:025003. [PMID: 25898400 DOI: 10.1088/1478-3975/12/2/025003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Lipid molecules, structural components of biomembranes, have been proposed for an important role in membrane fusion. Through various techniques based on a protein-reconstituted vesicle-vesicle fusion system, we investigated the influence of several lipid molecules on different stages of a yeast soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-mediated membrane fusion process. Lipid compositions played a significant role in the early stages, docking and lipid mixing, while only exhibiting a minor effect on fusion pore formation and dilation phases, indicated by both small and large content mixing.
Collapse
Affiliation(s)
- Ying Lai
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Molecular origins of synaptotagmin 1 activities on vesicle docking and fusion pore opening. Sci Rep 2015; 5:9267. [PMID: 25791821 PMCID: PMC4366854 DOI: 10.1038/srep09267] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/26/2015] [Indexed: 11/25/2022] Open
Abstract
Synaptotagmin 1 (Syt1), a major Ca2+ sensor in neuroexocytosis, utilizes SNARE- and membrane-binding to regulate vesicle fusion, a required process for neurotransmitter release at the synapse. However, the mechanism by which Syt1 orchestrates SNARE- and membrane- binding to control individual vesicle fusion steps is still unclear. In this study, we used a number of single vesicle assays that can differentiate intermediates of neuroexocytosis, to focus on Syt1 mutants that might impair Syt1-SNARE/PIP2 interaction, Ca2+-binding, or membrane penetration. Our results show that, although putative Syt1-SNARE/PIP2 coupling through the polybasic region of the C2B domain is critical for vesicle docking, its disruption does not affect content release. In contrast, Ca2+-binding and membrane-penetration mutants significantly reduce content release. Our results thus delineate multiple functions of Syt1 along the pathway of Ca2+-triggered exocytosis in unprecedented detail.
Collapse
|
65
|
Zhang Y, Diao J, Colbert KN, Lai Y, Pfuetzner RA, Padolina MS, Vivona S, Ressl S, Cipriano DJ, Choi UB, Shah N, Weis WI, Brunger AT. Munc18a does not alter fusion rates mediated by neuronal SNAREs, synaptotagmin, and complexin. J Biol Chem 2015; 290:10518-34. [PMID: 25716318 PMCID: PMC4400359 DOI: 10.1074/jbc.m114.630772] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Indexed: 01/25/2023] Open
Abstract
Sec1/Munc18 (SM) proteins are essential for membrane trafficking, but their molecular mechanism remains unclear. Using a single vesicle-vesicle content-mixing assay with reconstituted neuronal SNAREs, synaptotagmin-1, and complexin-1, we show that the neuronal SM protein Munc18a/nSec1 has no effect on the intrinsic kinetics of both spontaneous fusion and Ca2+-triggered fusion between vesicles that mimic synaptic vesicles and the plasma membrane. However, wild type Munc18a reduced vesicle association ∼50% when the vesicles bearing the t-SNAREs syntaxin-1A and SNAP-25 were preincubated with Munc18 for 30 min. Single molecule experiments with labeled SNAP-25 indicate that the reduction of vesicle association is a consequence of sequestration of syntaxin-1A by Munc18a and subsequent release of SNAP-25 (i.e. Munc18a captures syntaxin-1A via its high affinity interaction). Moreover, a phosphorylation mimic mutant of Munc18a with reduced affinity to syntaxin-1A results in less reduction of vesicle association. In summary, Munc18a does not directly affect fusion, although it has an effect on the t-SNARE complex, depending on the presence of other factors and experimental conditions. Our results suggest that Munc18a primarily acts at the prefusion stage.
Collapse
Affiliation(s)
- Yunxiang Zhang
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and
| | - Jiajie Diao
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and the Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| | - Karen N Colbert
- From the Departments of Molecular and Cellular Physiology, Structural Biology, and
| | - Ying Lai
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and
| | - Richard A Pfuetzner
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and the Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| | - Mark S Padolina
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and the Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| | - Sandro Vivona
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and
| | - Susanne Ressl
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and
| | - Daniel J Cipriano
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and the Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| | - Ucheor B Choi
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and the Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| | | | - William I Weis
- From the Departments of Molecular and Cellular Physiology, Structural Biology, and Photon Science and
| | - Axel T Brunger
- From the Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and the Howard Hughes Medical Institute, Stanford University, Stanford, California 94305
| |
Collapse
|
66
|
Lou X, Shin J, Yang Y, Kim J, Shin YK. Synaptotagmin-1 is an antagonist for Munc18-1 in SNARE zippering. J Biol Chem 2015; 290:10535-43. [PMID: 25716321 DOI: 10.1074/jbc.m114.631341] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Indexed: 12/17/2022] Open
Abstract
In neuroexocytosis, SNAREs and Munc18-1 may consist of the minimal membrane fusion machinery. Consistent with this notion, we observed, using single molecule fluorescence assays, that Munc18-1 stimulates SNARE zippering and SNARE-dependent lipid mixing in the absence of a major Ca(2+) sensor synaptotagmin-1 (Syt1), providing the structural basis for the conserved function of Sec1/Munc18 proteins in exocytosis. However, when full-length Syt1 is present, no enhancement of SNARE zippering and no acceleration of Ca(2+)-triggered content mixing by Munc18-1 are observed. Thus, our results show that Syt1 acts as an antagonist for Munc18-1 in SNARE zippering and fusion pore opening. Although the Sec1/Munc18 family may serve as part of the fusion machinery in other exocytotic pathways, Munc18-1 may have evolved to play a different role, such as regulating syntaxin-1a in neuroexocytosis.
Collapse
Affiliation(s)
- Xiaochu Lou
- From the Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa 50011 and
| | - Jaeil Shin
- From the Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa 50011 and
| | - Yoosoo Yang
- the Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Jaewook Kim
- the Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Yeon-Kyun Shin
- From the Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa 50011 and the Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| |
Collapse
|
67
|
Diao J, Liu R, Rong Y, Zhao M, Zhang J, Lai Y, Zhou Q, Wilz LM, Li J, Vivona S, Pfuetzner RA, Brunger AT, Zhong Q. ATG14 promotes membrane tethering and fusion of autophagosomes to endolysosomes. Nature 2015; 520:563-6. [PMID: 25686604 DOI: 10.1038/nature14147] [Citation(s) in RCA: 427] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 12/08/2014] [Indexed: 01/09/2023]
Abstract
Autophagy, an important catabolic pathway implicated in a broad spectrum of human diseases, begins by forming double membrane autophagosomes that engulf cytosolic cargo and ends by fusing autophagosomes with lysosomes for degradation. Membrane fusion activity is required for early biogenesis of autophagosomes and late degradation in lysosomes. However, the key regulatory mechanisms of autophagic membrane tethering and fusion remain largely unknown. Here we report that ATG14 (also known as beclin-1-associated autophagy-related key regulator (Barkor) or ATG14L), an essential autophagy-specific regulator of the class III phosphatidylinositol 3-kinase complex, promotes membrane tethering of protein-free liposomes, and enhances hemifusion and full fusion of proteoliposomes reconstituted with the target (t)-SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) syntaxin 17 (STX17) and SNAP29, and the vesicle (v)-SNARE VAMP8 (vesicle-associated membrane protein 8). ATG14 binds to the SNARE core domain of STX17 through its coiled-coil domain, and stabilizes the STX17-SNAP29 binary t-SNARE complex on autophagosomes. The STX17 binding, membrane tethering and fusion-enhancing activities of ATG14 require its homo-oligomerization by cysteine repeats. In ATG14 homo-oligomerization-defective cells, autophagosomes still efficiently form but their fusion with endolysosomes is blocked. Recombinant ATG14 homo-oligomerization mutants also completely lose their ability to promote membrane tethering and to enhance SNARE-mediated fusion in vitro. Taken together, our data suggest an autophagy-specific membrane fusion mechanism in which oligomeric ATG14 directly binds to STX17-SNAP29 binary t-SNARE complex on autophagosomes and primes it for VAMP8 interaction to promote autophagosome-endolysosome fusion.
Collapse
Affiliation(s)
- Jiajie Diao
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Rong Liu
- 1] Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA [2] Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA [3] College of Food Science &Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yueguang Rong
- 1] Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA [2] Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Minglei Zhao
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Jing Zhang
- 1] Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA [2] Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ying Lai
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Qiangjun Zhou
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Livia M Wilz
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | - Jianxu Li
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | - Sandro Vivona
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Richard A Pfuetzner
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Axel T Brunger
- 1] Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA [2] Department of Structural Biology, Stanford University, Stanford, California 94305, USA [3] Department of Photon Science, Stanford University, Stanford, California 94305, USA [4] Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, USA [5] Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Qing Zhong
- 1] Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA [2] Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
68
|
Aryani A, Denecke B. Exosomes as a Nanodelivery System: a Key to the Future of Neuromedicine? Mol Neurobiol 2014; 53:818-834. [PMID: 25502465 PMCID: PMC4752585 DOI: 10.1007/s12035-014-9054-5] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 12/05/2014] [Indexed: 01/04/2023]
Abstract
Since the beginning of the last decade, exosomes have been of increased interest in the science community. Exosomes represent a new kind of long distance transfer of biological molecules among cells. This review provides a comprehensive overview about the construction of exosomes, their targeting and their fusion mechanisms to the recipient cells. Complementarily, the current state of research regarding the cargo of exosomes is discussed. A particular focus was placed on the role of exosomes in the central nervous system. An increasing number of physiological processes in the brain could be associated with exosomes. In this context, it is becoming more apparent that exosomes are involved in several neurological and specifically neurodegenerative diseases. The treatment of these kinds of diseases is often difficult not least because of the blood-brain barrier. Exosomes are very stable, can pass the blood-brain barrier and, therefore, reveal bright perspectives towards diagnosis and therapeutic treatments. A prerequisite for clinical applications is a standardised approach. Features necessary for a standardised diagnosis using exosomes are discussed. In therapeutic terms, exosomes represent a promising drug delivery system able to pass the blood-brain barrier. One option to overcome the disadvantages potentially associated with the use of endogenous exosomes is the design of artificial exosomes. The artificial exosomes with a clearly defined therapeutic active cargo and surface marker ensuring the specific targeting to the recipient cells is proposed as a promising approach.
Collapse
Affiliation(s)
- Arian Aryani
- Interdisciplinary Center for Clinical Research Aachen (IZKF Aachen), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research Aachen (IZKF Aachen), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
69
|
Single molecule studies of RNA-RNA interactions. Methods Mol Biol 2014; 1240:97-112. [PMID: 25352139 DOI: 10.1007/978-1-4939-1896-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Non-coding RNAs including microRNAs, siRNAs, and snoRNAs interact with their targets directly through RNA-RNA interactions by base-paring (van Himbergen et al., Nucleic Acids Res 21(8):1713-1717, 1993). RNA-RNA interactions play important roles in gene transcription and protein translation, which can be investigated with several experimental techniques including single molecule methods. Here, we describe how single molecule Förster resonance energy transfer (FRET) can be used to study RNA-RNA interactions in vitro by either surface immobilization or vesicle encapsulation.
Collapse
|
70
|
Nanoscale high-content analysis using compositional heterogeneities of single proteoliposomes. Nat Methods 2014; 11:931-4. [PMID: 25086504 DOI: 10.1038/nmeth.3062] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 06/27/2014] [Indexed: 02/07/2023]
Abstract
Proteoliposome reconstitution is a standard method to stabilize purified transmembrane proteins in membranes for structural and functional assays. Here we quantified intrareconstitution heterogeneities in single proteoliposomes using fluorescence microscopy. Our results suggest that compositional heterogeneities can severely skew ensemble-average proteoliposome measurements but also enable ultraminiaturized high-content screens. We took advantage of this screening capability to map the oligomerization energy of the β2-adrenergic receptor using ∼10(9)-fold less protein than conventional assays.
Collapse
|
71
|
Analysis of interactions between SNARE proteins using imaging ellipsometer coupled with microfluidic array. Sci Rep 2014; 4:5341. [PMID: 24938428 PMCID: PMC4061542 DOI: 10.1038/srep05341] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 05/28/2014] [Indexed: 12/20/2022] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins are small and abundant membrane-bound proteins, whose specific interactions mediate membrane fusion during cell fusion or cellular trafficking. In this study, we report the use of a label-free method, called imaging ellipsometer to analyze the interactions among three SNAREs, namely Sec22p, Ykt6p and Sso2p. The SNAREs were immobilized on the silicon wafer and then analyzed in a pairwise mode with microfluidic array, leading us to discover the interactions between Ykt6p and Sso2p, Sec22p and Sso2p. Moreover, by using the real-time function of the imaging ellipsometer, we were able to obtain their association constants (KA) of about 104 M−1. We argue that the use of imaging ellipsometer coupled with microfluidic device will deepen our understanding of the molecular mechanisms underlying membrane fusion process.
Collapse
|
72
|
Synaptotagmin 1 and Ca2+ drive trans SNARE zippering. Sci Rep 2014; 4:4575. [PMID: 24694579 PMCID: PMC3974132 DOI: 10.1038/srep04575] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/17/2014] [Indexed: 12/21/2022] Open
Abstract
Synaptotagmin 1 (Syt1) is a major Ca2+-sensor that evokes neurotransmitter release. Here we used site-specific fluorescence resonance energy transfer (FRET) assay to investigate the effects of Syt1 on SNAREpin assembly. C2AB, a soluble version of Syt1, had virtually no stimulatory effect on the rate of the FRET at N-terminus of SNARE complex both with and without Ca2+, indicating C2AB does not interfere with the initial nucleation of SNARE assembly. However, C2AB-Ca2+ accelerated the FRET rate significantly at membrane proximal region, indicating C2AB-Ca2+ promotes the transition from a partially assembled SNARE complex to the fusion-competent SNAREpin. Similar enhancement was also observed at the end of the transmembrane domain of SNARE proteins. The stimulatory effect disappeared if there was no membrane or only neutral membrane present.
Collapse
|
73
|
Pantano S, Montecucco C. The blockade of the neurotransmitter release apparatus by botulinum neurotoxins. Cell Mol Life Sci 2014; 71:793-811. [PMID: 23749048 PMCID: PMC11113401 DOI: 10.1007/s00018-013-1380-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 12/22/2022]
Abstract
The high toxicity of the seven serotypes of botulinum neurotoxins (BoNT/A to G), together with their specificity and reversibility, includes them in the list A of potential bioterrorism weapons and, at the same time, among the therapeutics of choice for a variety of human syndromes. They invade nerve terminals and cleave specifically the three proteins which form the heterotrimeric SNAP REceptors (SNARE) complex that mediates neurotransmitter release. The BoNT-induced cleavage of the SNARE proteins explains by itself the paralysing activity of the BoNTs because the truncated proteins cannot form the SNARE complex. However, in the case of BoNT/A, the most widely used toxin in therapy, additional factors come into play as it only removes a few residues from the synaptosomal associate protein of 25 kDa C-terminus and this results in a long duration of action. To explain these facts and other experimental data, we present here a model for the assembly of the neuroexocytosis apparatus in which Synaptotagmin and Complexin first assist the zippering of the SNARE complex, and then stabilize and clamp an octameric radial assembly of the SNARE complexes.
Collapse
Affiliation(s)
- Sergio Pantano
- Institut Pasteur de Montevideo, Calle Mataojo 2020, CP 11400 Montevideo, Uruguay
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Institute of Neuroscience, National Research Council, Viale G. Colombo 3, 35121 Padua, Italy
| |
Collapse
|
74
|
Andrecka J, Spillane KM, Ortega-Arroyo J, Kukura P. Direct observation and control of supported lipid bilayer formation with interferometric scattering microscopy. ACS NANO 2013; 7:10662-70. [PMID: 24251388 DOI: 10.1021/nn403367c] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Supported lipid bilayers (SLB) are frequently used to study processes associated with or mediated by lipid membranes. The mechanism by which SLBs form is a matter of debate, largely due to the experimental difficulty associated with observing the adsorption and rupture of individual vesicles. Here, we used interferometric scattering microscopy (iSCAT) to directly visualize membrane formation from nanoscopic vesicles in real time. We observed a number of previously proposed phenomena such as vesicle adsorption, rupture, movement, and a wave-like bilayer spreading. By varying the vesicle size and the lipid-surface interaction strength, we rationalized and tuned the relative contributions of these phenomena to bilayer formation. Our results support a model where the interplay between bilayer edge tension and the overall interaction energy with the surface determine the mechanism of SLB formation. The unique combination of sensitivity, speed, and label-free imaging capability of iSCAT provides exciting prospects not only for investigations of SLB formation, but also for studies of assembly and disassembly processes on the nanoscale with previously unattainable accuracy and sensitivity.
Collapse
Affiliation(s)
- Joanna Andrecka
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford , South Parks Road, Oxford OX1 3QZ, United Kingdom
| | | | | | | |
Collapse
|
75
|
Chakraborty H, Tarafdar PK, Lentz BR. A novel assay for detecting fusion pore formation: implications for the fusion mechanism. Biochemistry 2013; 52:8510-7. [PMID: 24164461 DOI: 10.1021/bi401369j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Membrane fusion is broadly envisioned as a two- or three-step process proceeding from contacting bilayers through one or two semistable, nonlamellar lipidic intermediate structures to a fusion pore. A true fusion event requires mixing of contents between compartments and is monitored by the movement of soluble molecules between trapped compartments. We have used poly(ethylene glycol) (PEG) to rapidly generate an ensemble aggregated state A that proceeds sequentially through intermediates (I₁ and/or I₂) to a final fusion pore state (FP) with rate constants k₁, k₂, and k₃. Movement of moderately sized solutes (e.g., Tb³⁺/dipicolinic acid) has been used to detect pores assigned to intermediate states as well as to the final state (FP). Analysis of ensemble kinetic data has required that mixing of contents occurs with defined probabilities (αi) in each ensemble state, although it is unclear whether pores that form in different states are different. We introduce here a simple new assay that employs fluorescence resonance energy transfer (FRET) between a 6-carboxyfluorescein (donor) and tetramethylrhodamine (acceptor), which are covalently attached to complementary sequences of 10 bp oligonucleotides. Complementary sequences of fluorophore-labeled oligonucleotides were incorporated in vesicles separately, and the level of FRET increased in a simple exponential fashion during PEG-mediated fusion. The resulting rate constant corresponded closely to the slow rate constant of FP formation (k₃) derived from small molecule assays. Additionally, the total extent of oligonucleotide mixing corresponded to the fraction of content mixing that occurred in state FP in the small molecule assay. The results show that both large "final pores" and small (presumably transient) pores can form between vesicles throughout the fusion process. The implications of this result for the mechanism of membrane fusion are discussed.
Collapse
Affiliation(s)
- Hirak Chakraborty
- Department of Biochemistry and Biophysics and Program in Molecular and Cellular Biophysics, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599-7260, United States
| | | | | |
Collapse
|
76
|
Diao J, Cipriano DJ, Zhao M, Zhang Y, Shah S, Padolina MS, Pfuetzner RA, Brunger AT. Complexin-1 enhances the on-rate of vesicle docking via simultaneous SNARE and membrane interactions. J Am Chem Soc 2013; 135:15274-7. [PMID: 24083833 PMCID: PMC3854000 DOI: 10.1021/ja407392n] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
In
synaptic terminals, complexin is thought to have inhibitory
and activating roles for spontaneous “mini” release
and evoked synchronized neurotransmitter release, respectively. We
used single vesicle–vesicle microscopy imaging to study the
effect of complexin-1 on the on-rate of docking between vesicles that
mimic synaptic vesicles and the plasma membrane. We found that complexin-1
enhances the on-rate of docking of synaptic vesicle mimics containing
full-length synaptobrevin-2 and full-length synaptotagmin-1 to plasma
membrane-mimicking vesicles containing full-length syntaxin-1A and
SNAP-25A. This effect requires the C-terminal domain of complexin-1,
which binds to the membrane, the presence of PS in the membrane, and
the core region of complexin-1, which binds to the SNARE complex.
Collapse
Affiliation(s)
- Jiajie Diao
- Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, and Photon Science and Howard Hughes Medical Institute, Stanford University , Stanford, California 94305, United States
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Christensen AL, Lohr C, Christensen SM, Stamou D. Single vesicle biochips for ultra-miniaturized nanoscale fluidics and single molecule bioscience. LAB ON A CHIP 2013; 13:3613-3625. [PMID: 23856986 DOI: 10.1039/c3lc50492a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
One of the major bottlenecks in the development of biochips is maintaining the structure and function of biomolecules when interfacing them with hard matter (glass, plastics, metals, etc.), a challenge that is exacerbated during miniaturization that inevitably increases the interface to volume ratio of these devices. Biochips based on immobilized vesicles circumvent this problem by encapsulating biomolecules in the protective environment of a lipid bilayer, thus minimizing interactions with hard surfaces. Here we review the development of biochips based on arrays of single nanoscale vesicles, their fabrication via controlled self-assembly, and their characterization using fluorescence microscopy. We also highlight their applications in selected fields such as nanofluidics and single molecule bioscience. Despite their great potential for improved biocompatibility, extreme miniaturization and high throughput, single vesicle biochips are still a niche technology that has yet to establish its commercial relevance.
Collapse
Affiliation(s)
- Andreas L Christensen
- Bionanotechnology and Nanomedicine Laboratory, Department of Chemistry, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | | | | |
Collapse
|
78
|
Megighian A, Zordan M, Pantano S, Scorzeto M, Rigoni M, Zanini D, Rossetto O, Montecucco C. Evidence for a radial SNARE super-complex mediating neurotransmitter release at the Drosophila neuromuscular junction. J Cell Sci 2013; 126:3134-40. [PMID: 23687382 DOI: 10.1242/jcs.123802] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The SNARE proteins VAMP/synaptobrevin, SNAP-25 and syntaxin are core components of the apparatus that mediates neurotransmitter release. They form a heterotrimeric complex, and an undetermined number of SNARE complexes assemble to form a super-complex. Here, we present a radial model of this nanomachine. Experiments performed with botulinum neurotoxins led to the identification of one arginine residue in SNAP-25 and one aspartate residue in syntaxin (R206 and D253 in Drosophila melanogaster). These residues are highly conserved and predicted to play a major role in the protein-protein interactions between SNARE complexes by forming an ionic couple. Accordingly, we generated transgenic Drosophila lines expressing SNAREs mutated in these residues and performed an electrophysiological analysis of their neuromuscular junctions. Our results indicate that SNAP-25-R206 and syntaxin-D253 play a major role in neuroexocytosis and support a radial assembly of several SNARE complexes interacting via the ionic couple formed by these two residues.
Collapse
Affiliation(s)
- Aram Megighian
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 56 B, 35121 Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Diao J, Burré J, Vivona S, Cipriano DJ, Sharma M, Kyoung M, Südhof TC, Brunger AT. Native α-synuclein induces clustering of synaptic-vesicle mimics via binding to phospholipids and synaptobrevin-2/VAMP2. eLife 2013; 2:e00592. [PMID: 23638301 PMCID: PMC3639508 DOI: 10.7554/elife.00592] [Citation(s) in RCA: 241] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/25/2013] [Indexed: 12/13/2022] Open
Abstract
α-Synuclein is a presynaptic protein that is implicated in Parkinson's and other neurodegenerative diseases. Physiologically, native α-synuclein promotes presynaptic SNARE-complex assembly, but its molecular mechanism of action remains unknown. Here, we found that native α-synuclein promotes clustering of synaptic-vesicle mimics, using a single-vesicle optical microscopy system. This vesicle-clustering activity was observed for both recombinant and native α-synuclein purified from mouse brain. Clustering was dependent on specific interactions of native α-synuclein with both synaptobrevin-2/VAMP2 and anionic lipids. Out of the three familial Parkinson's disease-related point mutants of α-synuclein, only the lipid-binding deficient mutation A30P disrupted clustering, hinting at a possible loss of function phenotype for this mutant. α-Synuclein had little effect on Ca(2+)-triggered fusion in our reconstituted single-vesicle system, consistent with in vivo data. α-Synuclein may therefore lead to accumulation of synaptic vesicles at the active zone, providing a 'buffer' of synaptic vesicles, without affecting neurotransmitter release itself. DOI:http://dx.doi.org/10.7554/eLife.00592.001.
Collapse
Affiliation(s)
- Jiajie Diao
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
- Department of Structural Biology, Stanford University, Stanford, United States
- Departments of Photon Sciences, and Neurology and Neurological Sciences, Stanford University, Stanford, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Jacqueline Burré
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Sandro Vivona
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
- Department of Structural Biology, Stanford University, Stanford, United States
- Departments of Photon Sciences, and Neurology and Neurological Sciences, Stanford University, Stanford, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Daniel J Cipriano
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
- Department of Structural Biology, Stanford University, Stanford, United States
- Departments of Photon Sciences, and Neurology and Neurological Sciences, Stanford University, Stanford, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Manu Sharma
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Minjoung Kyoung
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
- Department of Structural Biology, Stanford University, Stanford, United States
- Departments of Photon Sciences, and Neurology and Neurological Sciences, Stanford University, Stanford, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, United States
- For correspondence: (TCS)
| | - Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
- Department of Structural Biology, Stanford University, Stanford, United States
- Departments of Photon Sciences, and Neurology and Neurological Sciences, Stanford University, Stanford, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, United States
- For correspondence: (ATB)
| |
Collapse
|
80
|
Diao J, Zhao M, Zhang Y, Kyoung M, Brunger AT. Studying protein-reconstituted proteoliposome fusion with content indicators in vitro. Bioessays 2013; 35:658-65. [PMID: 23625805 DOI: 10.1002/bies.201300010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In vitro reconstitution assays are commonly used to study biological membrane fusion. However, to date, most ensemble and single-vesicle experiments involving SNARE proteins have been performed only with lipid-mixing, but not content-mixing indicators. Through simultaneous detection of lipid and small content-mixing indicators, we found that lipid mixing often occurs seconds prior to content mixing, or without any content mixing at all, during a 50-seconds observation period, for Ca(2+) -triggered fusion with SNAREs, full-length synaptotagmin-1, and complexin. Our results illustrate the caveats of commonly used bulk lipid-mixing fusion experiments. We recommend that proteoliposome fusion experiments should always employ content-mixing indicators in addition to, or in place of, lipid-mixing indicators.
Collapse
Affiliation(s)
- Jiajie Diao
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
| | | | | | | | | |
Collapse
|
81
|
Otterstrom J, van Oijen AM. Visualization of membrane fusion, one particle at a time. Biochemistry 2013; 52:1654-68. [PMID: 23421412 DOI: 10.1021/bi301573w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Protein-mediated fusion between phospholipid bilayers is a fundamental and necessary mechanism for many cellular processes. The short-lived nature of the intermediate states visited during fusion makes it challenging to capture precise kinetic information using classical, ensemble-averaging biophysical techniques. Recently, a number of single-particle fluorescence microscopy-based assays that allow researchers to obtain highly quantitative data about the fusion process by observing individual fusion events in real time have been developed. These assays depend upon changes in the acquired fluorescence signal to provide a direct readout for transitions between the various fusion intermediates. The resulting data yield meaningful and detailed kinetic information about the transitory states en route to productive membrane fusion. In this review, we highlight recent in vitro and in vivo studies of membrane fusion at the single-particle level in the contexts of viral membrane fusion and SNARE-mediated synaptic vesicle fusion. These studies afford insight into mechanisms of coordination between fusion-mediating proteins as well as coordination of the overall fusion process with other cellular processes. The development of single-particle approaches to investigate membrane fusion and their successful application to a number of model systems have resulted in a new experimental paradigm and open up considerable opportunities to extend these methods to other biological processes that involve membrane fusion.
Collapse
Affiliation(s)
- Jason Otterstrom
- Harvard Biophysics Program, Harvard Medical School , 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | | |
Collapse
|
82
|
Fusion pore formation and expansion induced by Ca2+ and synaptotagmin 1. Proc Natl Acad Sci U S A 2013; 110:1333-8. [PMID: 23300284 DOI: 10.1073/pnas.1218818110] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fusion pore formation and expansion, crucial steps for neurotransmitter release and vesicle recycling in soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-dependent vesicle fusion, have not been well studied in vitro due to the lack of a reliable content-mixing fusion assay. Using methods detecting the intervesicular mixing of small and large cargoes at a single-vesicle level, we found that the neuronal SNARE complexes have the capacity to drive membrane hemifusion. However, efficient fusion pore formation and expansion require synaptotagmin 1 and Ca(2+). Real-time measurements show that pore expansion detected by content mixing of large DNA cargoes occurs much slower than initial pore formation that transmits small cargoes. Slow pore expansion perhaps provides a time window for vesicles to escape the full collapse fusion pathway via alternative mechanisms such as kiss-and-run. The results also show that complexin 1 stimulates pore expansion significantly, which could put bias between two pathways of vesicle recycling.
Collapse
|
83
|
Diao J, Grob P, Cipriano DJ, Kyoung M, Zhang Y, Shah S, Nguyen A, Padolina M, Srivastava A, Vrljic M, Shah A, Nogales E, Chu S, Brunger AT. Synaptic proteins promote calcium-triggered fast transition from point contact to full fusion. eLife 2012; 1:e00109. [PMID: 23240085 PMCID: PMC3514886 DOI: 10.7554/elife.00109] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 09/18/2012] [Indexed: 12/18/2022] Open
Abstract
The molecular underpinnings of synaptic vesicle fusion for fast neurotransmitter release are still unclear. Here, we used a single vesicle-vesicle system with reconstituted SNARE and synaptotagmin-1 proteoliposomes to decipher the temporal sequence of membrane states upon Ca(2+)-injection at 250-500 μM on a 100-ms timescale. Furthermore, detailed membrane morphologies were imaged with cryo-electron microscopy before and after Ca(2+)-injection. We discovered a heterogeneous network of immediate and delayed fusion pathways. Remarkably, all instances of Ca(2+)-triggered immediate fusion started from a membrane-membrane point-contact and proceeded to complete fusion without discernible hemifusion intermediates. In contrast, pathways that involved a stable hemifusion diaphragm only resulted in fusion after many seconds, if at all. When complexin was included, the Ca(2+)-triggered fusion network shifted towards the immediate pathway, effectively synchronizing fusion, especially at lower Ca(2+)-concentration. Synaptic proteins may have evolved to select this immediate pathway out of a heterogeneous network of possible membrane fusion pathways.DOI:http://dx.doi.org/10.7554/eLife.00109.001.
Collapse
Affiliation(s)
- Jiajie Diao
- Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Structural Biology, Photon Science and Howard Hughes Medical Institute , Stanford University , Stanford , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Studying calcium-triggered vesicle fusion in a single vesicle-vesicle content and lipid-mixing system. Nat Protoc 2012; 8:1-16. [PMID: 23222454 PMCID: PMC3566647 DOI: 10.1038/nprot.2012.134] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This protocol describes a single vesicle-vesicle microscopy system to study Ca(2+)-triggered vesicle fusion. Donor vesicles contain reconstituted synaptobrevin and synaptotagmin-1. Acceptor vesicles contain reconstituted syntaxin and synaptosomal-associated protein 25 (SNAP-25), and they are tethered to a PEG-coated glass surface. Donor vesicles are mixed with the tethered acceptor vesicles and incubated for several minutes at a zero-Ca(2+) concentration, resulting in a collection of single interacting vesicle pairs. The donor vesicles also contain two spectrally distinct fluorophores that allow simultaneous monitoring of temporal changes of the content and membrane. Upon Ca(2+) injection into the sample chamber, our system therefore differentiates between hemifusion and complete fusion of interacting vesicle pairs and determines the temporal sequence of these events on a sub-100-millisecond time scale. Other factors such as complexin can be easily added. Our system is unique in that it monitors both content and lipid mixing and starts from a metastable state of interacting vesicle pairs before Ca(2+) injection.
Collapse
|
85
|
Joo C, Fareh M, Kim VN. Bringing single-molecule spectroscopy to macromolecular protein complexes. Trends Biochem Sci 2012. [PMID: 23200186 DOI: 10.1016/j.tibs.2012.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Single-molecule fluorescence spectroscopy offers real-time, nanometer-resolution information. Over the past two decades, this emerging single-molecule technique has been rapidly adopted to investigate the structural dynamics and biological functions of proteins. Despite this remarkable achievement, single-molecule fluorescence techniques must be extended to macromolecular protein complexes that are physiologically more relevant for functional studies. In this review, we present recent major breakthroughs for investigating protein complexes within cell extracts using single-molecule fluorescence. We outline the challenges, future prospects and potential applications of these new single-molecule fluorescence techniques in biological and clinical research.
Collapse
Affiliation(s)
- Chirlmin Joo
- Kavli Institute of NanoScience, Department of BioNanoScience, Delft University of Technology, 2628CJ, Delft, The Netherlands.
| | | | | |
Collapse
|
86
|
Fusion of single proteoliposomes with planar, cushioned bilayers in microfluidic flow cells. Nat Protoc 2012; 7:903-20. [PMID: 22517259 DOI: 10.1038/nprot.2012.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Many biological processes rely on membrane fusion, and therefore assays to study its mechanisms are necessary. Here we report an assay with sensitivity to single-vesicle, and even to single-molecule events using fluorescently labeled vesicle-associated v-SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) liposomes and target-membrane-associated t-SNARE-reconstituted planar, supported bilayers (t-SBLs). Docking and fusion events can be detected using conventional far-field epifluorescence or total internal reflection fluorescence microscopy. In this assay, fusion is dependent on SNAP-25, one of the t-SNARE subunits that is required for fusion in vivo. The success of the assay is due to the use of: (i) bilayers covered with a thin layer of poly(ethylene glycol) (PEG) to control bilayer-bilayer and bilayer-substrate interactions, and (ii) microfluidic flow channels that present many advantages, such as the removal of nonspecifically bound liposomes by flow. The protocol takes 6-8 d to complete. Analysis can take up to 2 weeks.
Collapse
|