1
|
Tsemperouli M, Cheppali SK, Molina FR, Chetrit D, Landajuela A, Toomre D, Karatekin E. Vesicle docking and fusion pore modulation by the neuronal calcium sensor Synaptotagmin-1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612660. [PMID: 39314345 PMCID: PMC11419119 DOI: 10.1101/2024.09.12.612660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Synaptotagmin-1 (Syt1) is a major calcium sensor for rapid neurotransmitter release in neurons and hormone release in many neuroendocrine cells. It possesses two tandem cytosolic C2 domains that bind calcium, negatively charged phospholipids, and the neuronal SNARE complex. Calcium binding to Syt1 triggers exocytosis, but how this occurs is not well understood. Syt1 has additional roles in docking dense core vesicles (DCV) and synaptic vesicles (SV) to the plasma membrane (PM) and in regulating fusion pore dynamics. Thus, Syt1 perturbations could affect release through vesicle docking, fusion triggering, fusion pore regulation, or a combination of these. Here, using a human neuroendocrine cell line, we show that neutralization of highly conserved polybasic patches in either C2 domain of Syt1 impairs both DCV docking and efficient release of serotonin from DCVs. Interestingly, the same mutations resulted in larger fusion pores and faster release of serotonin during individual fusion events. Thus, Syt1's roles in vesicle docking, fusion triggering, and fusion pore control may be functionally related.
Collapse
Affiliation(s)
- Maria Tsemperouli
- Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT
- Nanobiology Institute, Yale University, West Haven, CT
| | - Sudheer Kumar Cheppali
- Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT
- Nanobiology Institute, Yale University, West Haven, CT
| | - Felix Rivera Molina
- Cell Biology, School of Medicine, Yale University
- CINEMA Lab, School of Medicine, Yale University
| | - David Chetrit
- Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT
- Nanobiology Institute, Yale University, West Haven, CT
| | - Ane Landajuela
- Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT
- Nanobiology Institute, Yale University, West Haven, CT
| | - Derek Toomre
- Cell Biology, School of Medicine, Yale University
- CINEMA Lab, School of Medicine, Yale University
| | - Erdem Karatekin
- Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, CT
- Nanobiology Institute, Yale University, West Haven, CT
- Molecular Biophysics and Biochemistry, Yale University, New Haven, CT
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université de Paris, Centre National de la Recherche Scientifique (CNRS) UMR 8003, Paris, France
- Wu Tsai Institute, Yale University
| |
Collapse
|
2
|
Shin KC, Ali Moussa HY, Park Y. Cholesterol imbalance and neurotransmission defects in neurodegeneration. Exp Mol Med 2024; 56:1685-1690. [PMID: 39085348 PMCID: PMC11371908 DOI: 10.1038/s12276-024-01273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 08/02/2024] Open
Abstract
The brain contains the highest concentration of cholesterol in the human body, which emphasizes the importance of cholesterol in brain physiology. Cholesterol is involved in neurogenesis and synaptogenesis, and age-related reductions in cholesterol levels can lead to synaptic loss and impaired synaptic plasticity, which potentially contribute to neurodegeneration. The maintenance of cholesterol homeostasis in the neuronal plasma membrane is essential for normal brain function, and imbalances in cholesterol distribution are associated with various neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, and Huntington's disease. This review aims to explore the molecular and pathological mechanisms by which cholesterol imbalance can lead to neurotransmission defects and neurodegeneration, focusing on four key mechanisms: (1) synaptic dysfunction, (2) alterations in membrane structure and protein clustering, (3) oligomers of amyloid beta (Aβ) protein, and (4) α-synuclein aggregation.
Collapse
Affiliation(s)
- Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Houda Yasmine Ali Moussa
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
- College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| |
Collapse
|
3
|
Patil SS, Sanghrajka K, Sriram M, Chakraborty A, Majumdar S, Bhaskar BR, Das D. Synaptobrevin2 monomers and dimers differentially engage to regulate the functional trans-SNARE assembly. Life Sci Alliance 2024; 7:e202402568. [PMID: 38238088 PMCID: PMC10796598 DOI: 10.26508/lsa.202402568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
The precise cell-to-cell communication relies on SNARE-catalyzed membrane fusion. Among ∼70 copies of synaptobrevin2 (syb2) in synaptic vesicles, only ∼3 copies are sufficient to facilitate the fusion process at the presynaptic terminal. It is unclear what dictates the number of SNARE complexes that constitute the fusion pore assembly. The structure-function relation of these dynamic pores is also unknown. Here, we demonstrate that syb2 monomers and dimers differentially engage in regulating the trans-SNARE assembly during membrane fusion. The differential recruitment of two syb2 structures at the membrane fusion site has consequences in regulating individual nascent fusion pore properties. We have identified a few syb2 transmembrane domain residues that control monomer/dimer conversion. Overall, our study indicates that syb2 monomers and dimers are differentially recruited at the release sites for regulating membrane fusion events.
Collapse
Affiliation(s)
- Swapnali S Patil
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Kinjal Sanghrajka
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Malavika Sriram
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Aritra Chakraborty
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sougata Majumdar
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Bhavya R Bhaskar
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Debasis Das
- https://ror.org/03ht1xw27 Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
4
|
Perpiñá-Clérigues C, Mellado S, Galiana-Roselló C, Fernández-Regueras M, Marcos M, García-García F, Pascual M. Novel insight into the lipid network of plasma extracellular vesicles reveal sex-based differences in the lipidomic profile of alcohol use disorder patients. Biol Sex Differ 2024; 15:10. [PMID: 38273378 PMCID: PMC10809459 DOI: 10.1186/s13293-024-00584-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Alcohol use disorder (AUD) is one of the most common psychiatric disorders, with the consumption of alcohol considered a leading cause of preventable deaths worldwide. Lipids play a crucial functional role in cell membranes; however, we know little about the role of lipids in extracellular vesicles (EVs) as regulatory molecules and disease biomarkers. METHODS We employed a sensitive lipidomic strategy to characterize lipid species from the plasma EVs of AUD patients to evaluate functional roles and enzymatic activity networks to improve the knowledge of lipid metabolism after alcohol consumption. We analyzed plasma EV lipids from AUD females and males and healthy individuals to highlight lipids with differential abundance and biologically interpreted lipidomics data using LINEX2, which evaluates enzymatic dysregulation using an enrichment algorithm. RESULTS Our results show, for the first time, that AUD females exhibited more significant substrate-product changes in lysophosphatidylcholine/phosphatidylcholine lipids and phospholipase/acyltransferase activity, which are potentially linked to cancer progression and neuroinflammation. Conversely, AUD males suffer from dysregulated ceramide and sphingomyelin lipids involving sphingomyelinase, sphingomyelin phosphodiesterase, and sphingomyelin synthase activity, which relates to hepatotoxicity. Notably, the analysis of plasma EVs from AUD females and males demonstrates enrichment of lipid ontology terms associated with "negative intrinsic curvature" and "positive intrinsic curvature", respectively. CONCLUSIONS Our methodological developments support an improved understanding of lipid metabolism and regulatory mechanisms, which contribute to the identification of novel lipid targets and the discovery of sex-specific clinical biomarkers in AUD.
Collapse
Affiliation(s)
- Carla Perpiñá-Clérigues
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Cristina Galiana-Roselló
- Department of Inorganic Chemistry, Institute of Molecular Science, University of Valencia, 46980, Paterna, Spain
| | - María Fernández-Regueras
- Hospital Universitario de Burgos, 09006, Burgos, Spain
- Hospital Universitario de Salamanca, 37007, Salamanca, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Francisco García-García
- Computational Biomedicine Laboratory, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain.
| |
Collapse
|
5
|
Su R, Wang S, McDargh Z, O'Shaughnessy B. Three membrane fusion pore families determine the pathway to pore dilation. Biophys J 2023; 122:3986-3998. [PMID: 37644721 PMCID: PMC10560699 DOI: 10.1016/j.bpj.2023.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/19/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
During exocytosis secretory vesicles fuse with a target membrane and release neurotransmitters, hormones, or other bioactive molecules through a membrane fusion pore. The initially small pore may subsequently dilate for full contents release, as commonly observed in amperometric traces. The size, shape, and evolution of the pore is critical to the course of contents release, but exact fusion pore solutions accounting for membrane tension and bending energy constraints have not been available. Here, we obtained exact solutions for fusion pores between two membranes. We find three families: a narrow pore, a wide pore, and an intermediate tether-like pore. For high tensions these are close to the catenoidal and tether solutions recently reported for freely hinged membrane boundaries. We suggest membrane fusion initially generates a stable narrow pore, and the dilation pathway is a transition to the stable wide pore family. The unstable intermediate pore is the transition state that sets the energy barrier for this dilation pathway. Pore dilation is mechanosensitive, as the energy barrier is lowered by increased membrane tension. Finally, we study fusion pores in nanodiscs, powerful systems for the study of individual pores. We show that nanodiscs stabilize fusion pores by locking them into the narrow pore family.
Collapse
Affiliation(s)
- Rui Su
- Department of Chemical Engineering, Columbia University, New York City, New York
| | - Shuyuan Wang
- Department of Chemical Engineering, Columbia University, New York City, New York; Department of Physics, Columbia University, New York City, New York
| | - Zachary McDargh
- Department of Chemical Engineering, Columbia University, New York City, New York
| | - Ben O'Shaughnessy
- Department of Chemical Engineering, Columbia University, New York City, New York.
| |
Collapse
|
6
|
Golani G, Schwarz US. High curvature promotes fusion of lipid membranes: Predictions from continuum elastic theory. Biophys J 2023; 122:1868-1882. [PMID: 37077047 PMCID: PMC10209146 DOI: 10.1016/j.bpj.2023.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/19/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023] Open
Abstract
The fusion of lipid membranes progresses through a series of hemifusion intermediates with two significant energy barriers related to the formation of stalk and fusion pore, respectively. These energy barriers determine the speed and success rate of many critical biological processes, including the fusion of highly curved membranes, for example synaptic vesicles and enveloped viruses. Here we use continuum elastic theory of lipid monolayers to determine the relationship between membrane shape and energy barriers to fusion. We find that the stalk formation energy decreases with curvature by up to 31 kBT in a 20-nm-radius vesicle compared with planar membranes and by up to 8 kBT in the fusion of highly curved, long, tubular membranes. In contrast, the fusion pore formation energy barrier shows a more complicated behavior. Immediately after stalk expansion to the hemifusion diaphragm, the fusion pore formation energy barrier is low (15-25 kBT) due to lipid stretching in the distal monolayers and increased tension in highly curved vesicles. Therefore, the opening of the fusion pore is faster. However, these stresses relax over time due to lipid flip-flop from the proximal monolayer, resulting in a larger hemifusion diaphragm and a higher fusion pore formation energy barrier, up to 35 kBT. Therefore, if the fusion pore fails to open before significant lipid flip-flop takes place, the reaction proceeds to an extended hemifusion diaphragm state, which is a dead-end configuration in the fusion process and can be used to prevent viral infections. In contrast, in the fusion of long tubular compartments, the surface tension does not accumulate due to the formation of the diaphragm, and the energy barrier for pore expansion increases with curvature by up to 11 kBT. This suggests that inhibition of polymorphic virus infection could particularly target this feature of the second barrier.
Collapse
Affiliation(s)
- Gonen Golani
- Institute for Theoretical Physics and BioQuant Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Ulrich S Schwarz
- Institute for Theoretical Physics and BioQuant Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
7
|
Ali Moussa HY, Shin KC, Ponraj J, Kim SJ, Ryu J, Mansour S, Park Y. Requirement of Cholesterol for Calcium-Dependent Vesicle Fusion by Strengthening Synaptotagmin-1-Induced Membrane Bending. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206823. [PMID: 37058136 PMCID: PMC10214243 DOI: 10.1002/advs.202206823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/09/2023] [Indexed: 05/27/2023]
Abstract
Cholesterol is essential for neuronal activity and function. Cholesterol depletion in the plasma membrane impairs synaptic transmission. However, the molecular mechanisms by which cholesterol deficiency leads to defects in vesicle fusion remain poorly understood. Here, it is shown that cholesterol is required for Ca2+ -dependent native vesicle fusion using the in vitro reconstitution of fusion and amperometry to monitor exocytosis in chromaffin cells. Purified native vesicles are crucial for the reconstitution of physiological Ca2+ -dependent fusion, because vesicle-mimicking liposomes fail to reproduce the cholesterol effect. Intriguingly, cholesterol has no effect on the membrane binding of synaptotagmin-1, a Ca2+ sensor for ultrafast fusion. Cholesterol strengthens local membrane deformation and bending induced by synaptotagmin-1, thereby lowering the energy barrier for Ca2+ -dependent fusion to occur. The data provide evidence that cholesterol depletion abolishes Ca2+ -dependent vesicle fusion by disrupting synaptotagmin-1-induced membrane bending, and suggests that cholesterol is an essential lipid regulator for Ca2+ -dependent fusion.
Collapse
Affiliation(s)
- Houda Yasmine Ali Moussa
- Neurological Disorders Research CenterQatar Biomedical Research Institute (QBRI)Hamad Bin Khalifa University (HBKU)Qatar FoundationDohaQatar
| | - Kyung Chul Shin
- Neurological Disorders Research CenterQatar Biomedical Research Institute (QBRI)Hamad Bin Khalifa University (HBKU)Qatar FoundationDohaQatar
| | | | - Soo Jin Kim
- Division of Molecular and Life SciencesPohang University of Science and TechnologyPohang790‐784Republic of Korea
| | - Je‐Kyung Ryu
- Department of Physics & AstronomySeoul National University. 1 Gwanak‐roGwanak‐guSeoul08826South Korea
| | - Said Mansour
- HBKU Core LabsHamad Bin Khalifa University (HBKU)DohaQatar
| | - Yongsoo Park
- Neurological Disorders Research CenterQatar Biomedical Research Institute (QBRI)Hamad Bin Khalifa University (HBKU)Qatar FoundationDohaQatar
- College of Health & Life Sciences (CHLS)Hamad Bin Khalifa University (HBKU)Qatar FoundationDohaQatar
| |
Collapse
|
8
|
Mahmood A, Otruba Z, Weisgerber AW, Palay MD, Nguyen MT, Bills BL, Knowles MK. Exosome secretion kinetics are controlled by temperature. Biophys J 2023; 122:1301-1314. [PMID: 36814381 PMCID: PMC10111348 DOI: 10.1016/j.bpj.2023.02.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/12/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
When multivesicular endosomes (MVEs) fuse with the plasma membrane, exosomes are released into the extracellular space where they can affect other cells. The ability of exosomes to regulate cells nearby or further away depends on whether they remain attached to the secreting cell membrane. The regulation and kinetics of exosome secretion are not well characterized, but probes for directly imaging single MVE fusion events have allowed for visualization of the fusion and release process. In particular, the design of an exosome marker with a pH-sensitive dye in the middle of the tetraspanin protein CD63 has facilitated studies of individual MVE fusion events. Using TIRF microscopy, single fusion events were measured in A549 cells held at 23-37°C and events were identified using an automated detection algorithm. Stable docking precedes fusion almost always and a decrease in temperature was accompanied by decrease in the rate of content loss and in the frequency of fusion events. The loss of CD63-pHluorin fluorescence was measured at fusion sites and fit with a single or double exponential decay, with most events requiring two components and a plateau because the loss of fluorescence was typically incomplete. To interpret the kinetics, fusion events were simulated as a localized release of tethered/untethered exosomes coupled with the membrane diffusion of CD63. The experimentally observed decay required three components in the simulation: 1) free exosomes, 2) CD63 membrane diffusion from the endosomal membrane into the plasma membrane, and 3) tethered exosomes. Modeling with slow diffusion of the tethered exosomes (0.0015-0.004 μm2/s) accurately fits the experimental data for all temperatures. However, simulating with immobile tethers or the absence of tethers fails to replicate the data. Our model suggests that exosome release from the fusion site is incomplete due to postfusion, membrane attachment.
Collapse
Affiliation(s)
- Anarkali Mahmood
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| | - Zdeněk Otruba
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| | - Alan W Weisgerber
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| | - Max D Palay
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| | - Melodie T Nguyen
- Molecular and Cellular Biophysics Program, University of Denver, Denver, Colorado
| | - Broderick L Bills
- Molecular and Cellular Biophysics Program, University of Denver, Denver, Colorado
| | - Michelle K Knowles
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado; Molecular and Cellular Biophysics Program, University of Denver, Denver, Colorado.
| |
Collapse
|
9
|
Chen PC, Wang CT. Rat Pheochromocytoma PC12 Cells in Culture. Methods Mol Biol 2023; 2565:3-15. [PMID: 36205883 DOI: 10.1007/978-1-0716-2671-9_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] [Indexed: 06/16/2023]
Abstract
PC12 cells serve as a secretory cell model, especially suitable for studying the molecular mechanisms underlying fusion pore kinetics in regulated exocytosis of dense-core vesicles (DCVs). In this chapter, we describe a series of PC12 cell culture procedures optimized for real-time functional assays such as single-vesicle amperometry. In addition, these conditions have been widely used for single-cell biochemical assays such as the proximity ligation assay with immunostaining.
Collapse
Affiliation(s)
- Pin-Chun Chen
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Chih-Tien Wang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Yan C, Jiang J, Yang Y, Geng X, Dong W. The function of VAMP2 in mediating membrane fusion: An overview. Front Mol Neurosci 2022; 15:948160. [PMID: 36618823 PMCID: PMC9816800 DOI: 10.3389/fnmol.2022.948160] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Vesicle-associated membrane protein 2 (VAMP2, also known as synaptobrevin-2), encoded by VAMP2 in humans, is a key component of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex. VAMP2 combined with syntaxin-1A (SYX-1A) and synaptosome-associated protein 25 (SNAP-25) produces a force that induces the formation of fusion pores, thereby mediating the fusion of synaptic vesicles and the release of neurotransmitters. VAMP2 is largely unstructured in the absence of interaction partners. Upon interaction with other SNAREs, the structure of VAMP2 stabilizes, resulting in the formation of four structural domains. In this review, we highlight the current knowledge of the roles of the VAMP2 domains and the interaction between VAMP2 and various fusion-related proteins in the presynaptic cytoplasm during the fusion process. Our summary will contribute to a better understanding of the roles of the VAMP2 protein in membrane fusion.
Collapse
Affiliation(s)
- Chong Yan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Jie Jiang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqi Geng
- Department of Neurosurgery, Neurosurgical Clinical Research Center of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, China,*Correspondence: Xiaoqi Geng,
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China,Wei Dong,
| |
Collapse
|
11
|
Villamil Giraldo AM, Mannsverk S, Kasson PM. Measuring single-virus fusion kinetics using an assay for nucleic acid exposure. Biophys J 2022; 121:4467-4475. [PMID: 36330566 PMCID: PMC9748363 DOI: 10.1016/j.bpj.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/26/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
The kinetics by which individual enveloped viruses fuse with membranes provide an important window into viral-entry mechanisms. We have developed a real-time assay using fluorescent probes for single-virus genome exposure than can report on stages of viral entry including or subsequent to fusion pore formation and prior to viral genome trafficking. We accomplish this using oxazole yellow nucleic-acid-binding dyes, which can be encapsulated in the lumen of target membranes to permit specific detection of fusion events. Since increased fluorescence of the dye occurs only when it encounters viral genome via a fusion pore and binds, this assay excludes content leakage without fusion. Using this assay, we show that influenza virus fuses with liposomes of different sizes with indistinguishable kinetics by both testing liposomes extruded through pores of different radii and showing that the fusion kinetics of individual liposomes are uncorrelated with the size of the liposome. These results suggest that the starting curvature of such liposomes does not control the rate-limiting steps in influenza entry.
Collapse
Affiliation(s)
- Ana M Villamil Giraldo
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Steinar Mannsverk
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Peter M Kasson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden; Departments of Molecular Physiology and Biomedical Engineering, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
12
|
Chang CW, Hsiao YT, Scheuer KS, Jackson MB. Full-Fusion and Kiss-and-Run in Chromaffin Cells controlled by Irreversible Vesicle Size-Dependent Fusion Pore Transitions. Cell Calcium 2022; 105:102606. [DOI: 10.1016/j.ceca.2022.102606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/18/2022] [Indexed: 11/02/2022]
|
13
|
Nikolaus J, Hancock K, Tsemperouli M, Baddeley D, Karatekin E. Optimal Detection of Fusion Pore Dynamics Using Polarized Total Internal Reflection Fluorescence Microscopy. Front Mol Biosci 2021; 8:740408. [PMID: 34859048 PMCID: PMC8631473 DOI: 10.3389/fmolb.2021.740408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
The fusion pore is the initial narrow connection that forms between fusing membranes. During vesicular release of hormones or neurotransmitters, the nanometer-sized fusion pore may open-close repeatedly (flicker) before resealing or dilating irreversibly, leading to kiss-and-run or full-fusion events, respectively. Pore dynamics govern vesicle cargo release and the mode of vesicle recycling, but the mechanisms are poorly understood. This is partly due to a lack of reconstituted assays that combine single-pore sensitivity and high time resolution. Total internal reflection fluorescence (TIRF) microscopy offers unique advantages for characterizing single membrane fusion events, but signals depend on effects that are difficult to disentangle, including the polarization of the excitation electric field, vesicle size, photobleaching, orientation of the excitation dipoles of the fluorophores with respect to the membrane, and the evanescent field depth. Commercial TIRF microscopes do not allow control of excitation polarization, further complicating analysis. To overcome these challenges, we built a polarization-controlled total internal reflection fluorescence (pTIRF) microscope and monitored fusion of proteoliposomes with planar lipid bilayers with single molecule sensitivity and ∼15 ms temporal resolution. Using pTIRF microscopy, we detected docking and fusion of fluorescently labeled small unilamellar vesicles, reconstituted with exocytotic/neuronal v-SNARE proteins (vSUVs), with a supported bilayer containing the cognate t-SNAREs (tSBL). By varying the excitation polarization angle, we were able to identify a dye-dependent optimal polarization at which the fluorescence increase upon fusion was maximal, facilitating event detection and analysis of lipid transfer kinetics. An improved algorithm allowed us to estimate the size of the fusing vSUV and the fusion pore openness (the fraction of time the pore is open) for every event. For most events, lipid transfer was much slower than expected for diffusion through an open pore, suggesting that fusion pore flickering limits lipid release. We find a weak correlation between fusion pore openness and vesicle area. The approach can be used to study mechanisms governing fusion pore dynamics in a wide range of membrane fusion processes.
Collapse
Affiliation(s)
- Joerg Nikolaus
- Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
- Nanobiology Institute, Yale University, West Haven, CT, United States
| | - Kasey Hancock
- Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
- Nanobiology Institute, Yale University, West Haven, CT, United States
- Integrated Physical and Engineering Biology Program, Yale University, New Haven, CT, United States
| | - Maria Tsemperouli
- Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
- Nanobiology Institute, Yale University, West Haven, CT, United States
| | - David Baddeley
- Nanobiology Institute, Yale University, West Haven, CT, United States
- Cell Biology, Yale University, New Haven, CT, United States
| | - Erdem Karatekin
- Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
- Nanobiology Institute, Yale University, West Haven, CT, United States
- Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, United States
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université de Paris, Centre National de la Recherche Scientifique (CNRS), Paris, France
| |
Collapse
|
14
|
Flagging fusion: Phosphatidylserine signaling in cell-cell fusion. J Biol Chem 2021; 296:100411. [PMID: 33581114 PMCID: PMC8005811 DOI: 10.1016/j.jbc.2021.100411] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Formations of myofibers, osteoclasts, syncytiotrophoblasts, and fertilized zygotes share a common step, cell–cell fusion. Recent years have brought about considerable progress in identifying some of the proteins involved in these and other cell-fusion processes. However, even for the best-characterized cell fusions, we still do not know the mechanisms that regulate the timing of cell-fusion events. Are they fully controlled by the expression of fusogenic proteins or do they also depend on some triggering signal that activates these proteins? The latter scenario would be analogous to the mechanisms that control the timing of exocytosis initiated by Ca2+ influx and virus-cell fusion initiated by low pH- or receptor interaction. Diverse cell fusions are accompanied by the nonapoptotic exposure of phosphatidylserine at the surface of fusing cells. Here we review data on the dependence of membrane remodeling in cell fusion on phosphatidylserine and phosphatidylserine-recognizing proteins and discuss the hypothesis that cell surface phosphatidylserine serves as a conserved “fuse me” signal regulating the time and place of cell-fusion processes.
Collapse
|
15
|
Wu L, Courtney KC, Chapman ER. Cholesterol stabilizes recombinant exocytic fusion pores by altering membrane bending rigidity. Biophys J 2021; 120:1367-1377. [PMID: 33582136 DOI: 10.1016/j.bpj.2021.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/18/2021] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
SNARE-mediated membrane fusion proceeds via the formation of a fusion pore. This intermediate structure is highly dynamic and can flicker between open and closed states. In cells, cholesterol has been reported to affect SNARE-mediated exocytosis and fusion pore dynamics. Here, we address the question of whether cholesterol directly affects the flickering rate of reconstituted fusion pores in vitro. These experiments were enabled by the recent development of a nanodisc⋅black lipid membrane recording system that monitors dynamic transitions between the open and closed states of nascent recombinant pores with submillisecond time resolution. The fusion pores formed between nanodiscs that bore the vesicular SNARE synaptobrevin 2 and black lipid membranes that harbored the target membrane SNAREs syntaxin 1A and SNAP-25B were markedly affected by cholesterol. These effects include strong reductions in flickering out of the open state, resulting in a significant increase in the open dwell-time. We attributed these effects to the known role of cholesterol in altering the elastic properties of lipid bilayers because manipulation of phospholipids to increase membrane stiffness mirrored the effects of cholesterol. In contrast to the observed effects on pore kinetics, cholesterol had no effect on the current that passed through individual pores and, hence, did not affect pore size. In conclusion, our results show that cholesterol dramatically stabilizes fusion pores in the open state by increasing membrane bending rigidity.
Collapse
Affiliation(s)
- Lanxi Wu
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, Madison, Wisconsin
| | - Kevin C Courtney
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, Madison, Wisconsin
| | - Edwin R Chapman
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, Madison, Wisconsin.
| |
Collapse
|
16
|
Rahman MM, Williams SJ. Membrane tension may define the deadliest virus infection. COLLOID AND INTERFACE SCIENCE COMMUNICATIONS 2021; 40:100338. [PMID: 34722169 PMCID: PMC8544801 DOI: 10.1016/j.colcom.2020.100338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 05/23/2023]
Abstract
This manuscript describes the potentially significant role of interfacial tension in viral infection. Our hypothesis is based on evidence from drop coalescence hydrodynamics. A change in membrane tension can trigger fusion between the vesicle and cell such that genetic material, like viral RNA, can subsequently be transported to the cell interior. In other cases, RNA may reside near the cell membrane inside the cell, which could make their removal energetically unfavorable because of hydrodynamic interactions between membrane and RNA. Interfacial tension of the virus membrane can be modulated by temperature, among many other factors, of the mucosa layer. We discuss our hypothesis within the scope of recent SARS-CoV-2 studies where temperature-dependent membrane surface tension could be impacted through different atmospheric conditions, air conditioning systems, and the use of masks.
Collapse
Affiliation(s)
| | - Stuart J Williams
- Department of Mechanical Engineering, University of Louisville, KY, USA
| |
Collapse
|
17
|
He X, Ewing AG. Counteranions in the Stimulation Solution Alter the Dynamics of Exocytosis Consistent with the Hofmeister Series. J Am Chem Soc 2020; 142:12591-12595. [PMID: 32598145 PMCID: PMC7386575 DOI: 10.1021/jacs.0c05319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
![]()
We
show that the Hofmeister series of ions can be used to explain
the cellular changes in exocytosis observed by single-cell amperometry
for different counteranions. The formation, expansion, and closing
of the membrane fusion pore during exocytosis was found to be strongly
dependent on the counteranion species in solution. With stimulation
of chaotropic anions (e.g., ClO4–), the
expansion and closing time of the fusion pore are longer, suggesting
chaotropes can extend the duration of exocytosis compared with kosmotropic
anions (e.g., Cl–). At a concentration of 30 mM,
the two parameters (e.g., t1/2 and tfall) that define the duration of exocytosis
vary with the Hofmeister series (Cl– < Br– < NO3– ≤ ClO4– < SCN–). More interestingly,
fewer (e.g., Nfoot/Nevents) and smaller (e.g., Ifoot) prespike events are observed when chaotropes are counterions in
the stimulation solution, and the values can be sorted by the reverse
Hofmeister series (Cl– ≥ Br– > NO3– > ClO4– > SCN–). Based on ion specificity,
an adsorption-repulsion
mechanism, we suggest that the exocytotic Hofmeister series effect
originates from a looser swelling lipid bilayer structure due to the
adsorption and electrostatic repulsion of chaotropes on the hydrophobic
portion of the membrane. Our results provide a chemical link between
the Hofmeister series and the cellular process of neurotransmitter
release via exocytosis and provide a better physical framework to
understand this important phenomenon.
Collapse
Affiliation(s)
- Xiulan He
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Andrew G Ewing
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| |
Collapse
|
18
|
Dhara M, Mantero Martinez M, Makke M, Schwarz Y, Mohrmann R, Bruns D. Synergistic actions of v-SNARE transmembrane domains and membrane-curvature modifying lipids in neurotransmitter release. eLife 2020; 9:e55152. [PMID: 32391794 PMCID: PMC7239655 DOI: 10.7554/elife.55152] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023] Open
Abstract
Vesicle fusion is mediated by assembly of SNARE proteins between opposing membranes. While previous work suggested an active role of SNARE transmembrane domains (TMDs) in promoting membrane merger (Dhara et al., 2016), the underlying mechanism remained elusive. Here, we show that naturally-occurring v-SNARE TMD variants differentially regulate fusion pore dynamics in mouse chromaffin cells, indicating TMD flexibility as a mechanistic determinant that facilitates transmitter release from differentially-sized vesicles. Membrane curvature-promoting phospholipids like lysophosphatidylcholine or oleic acid profoundly alter pore expansion and fully rescue the decelerated fusion kinetics of TMD-rigidifying VAMP2 mutants. Thus, v-SNARE TMDs and phospholipids cooperate in supporting membrane curvature at the fusion pore neck. Oppositely, slowing of pore kinetics by the SNARE-regulator complexin-2 withstands the curvature-driven speeding of fusion, indicating that pore evolution is tightly coupled to progressive SNARE complex formation. Collectively, TMD-mediated support of membrane curvature and SNARE force-generated membrane bending promote fusion pore formation and expansion.
Collapse
Affiliation(s)
- Madhurima Dhara
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland UniversityHomburgGermany
| | - Maria Mantero Martinez
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland UniversityHomburgGermany
| | - Mazen Makke
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland UniversityHomburgGermany
| | - Yvonne Schwarz
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland UniversityHomburgGermany
| | - Ralf Mohrmann
- Institute for Physiology, Otto-von-Guericke UniversityMagdeburgGermany
| | - Dieter Bruns
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland UniversityHomburgGermany
| |
Collapse
|
19
|
Sahu A, Glisman A, Tchoufag J, Mandadapu KK. Geometry and dynamics of lipid membranes: The Scriven-Love number. Phys Rev E 2020; 101:052401. [PMID: 32575240 DOI: 10.1103/physreve.101.052401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/07/2020] [Indexed: 06/11/2023]
Abstract
The equations governing lipid membrane dynamics in planar, spherical, and cylindrical geometries are presented here. Unperturbed and first-order perturbed equations are determined and nondimensionalized. In membrane systems with a nonzero base flow, perturbed in-plane and out-of-plane quantities are found to vary over different length scales. A new dimensionless number, named the Scriven-Love number, and the well-known Föppl-von Kármán number result from a scaling analysis. The Scriven-Love number compares out-of-plane forces arising from the in-plane, intramembrane viscous stresses to the familiar elastic bending forces, while the Föppl-von Kármán number compares tension to bending forces. Both numbers are calculated in past experimental works, and span a wide range of values in various biological processes across different geometries. In situations with large Scriven-Love and Föppl-von Kármán numbers, the dynamical response of a perturbed membrane is dominated by out-of-plane viscous and surface tension forces-with bending forces playing a negligible role. Calculations of non-negligible Scriven-Love numbers in various biological processes and in vitro experiments show in-plane intramembrane viscous flows cannot generally be ignored when analyzing lipid membrane behavior.
Collapse
Affiliation(s)
- Amaresh Sahu
- Department of Chemical & Biomolecular Engineering, University of California, Berkeley, California 94720, USA
| | - Alec Glisman
- Department of Chemical & Biomolecular Engineering, University of California, Berkeley, California 94720, USA
| | - Joël Tchoufag
- Department of Chemical & Biomolecular Engineering, University of California, Berkeley, California 94720, USA
| | - Kranthi K Mandadapu
- Department of Chemical & Biomolecular Engineering, University of California, Berkeley, California 94720, USA
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| |
Collapse
|
20
|
Varga K, Jiang ZJ, Gong LW. Phosphatidylserine is critical for vesicle fission during clathrin-mediated endocytosis. J Neurochem 2019; 152:48-60. [PMID: 31587282 DOI: 10.1111/jnc.14886] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Phosphatidylserine (PS), a negatively charged phospholipid present predominantly at the inner leaflet of the plasma membrane, has been widely implicated in many cellular processes including membrane trafficking. Along this line, PS has been demonstrated to be important for endocytosis, however, the involved mechanisms remain uncertain. By monitoring clathrin-mediated endocytosis (CME) of single vesicles in mouse chromaffin cells using cell-attached capacitance measurements that offer millisecond time resolution, we demonstrate in the present study that the fission-pore duration is reduced by PS addition, indicating a stimulatory role of PS in regulating the dynamics of vesicle fission during CME. Furthermore, our results show that the PS-mediated effect on the fission-pore duration is Ca2+ -dependent and abolished in the absence of synaptotagmin 1 (Syt1), implying that Syt1 is necessary for the stimulatory role of PS in vesicle fission during CME. Consistently, a Syt1 mutant with a defective PS-Syt1 interaction increases the fission-pore duration. Taken together, our study suggests that PS-Syt1 interaction may be critical in regulating fission dynamics during CME.
Collapse
Affiliation(s)
- Kelly Varga
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Biological Sciences, University of North Texas at Dallas, Dallas, Texas, USA
| | - Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
21
|
Holz RW, Zimmerberg J. Dynamic Relationship of the SNARE Complex with a Membrane. Biophys J 2019; 117:627-630. [PMID: 31378313 DOI: 10.1016/j.bpj.2019.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/19/2019] [Accepted: 07/09/2019] [Indexed: 11/28/2022] Open
Abstract
Fusion of secretory granules and synaptic vesicles with the plasma membrane is driven by SNARE protein interactions. Intensive investigations in vitro, which include x-ray crystallography, cryoelectron microscopy, and NMR analyses by numerous groups, have elucidated structures relevant to the function of these proteins. Although function depends on the proteins being membrane bound, for experimental reasons, most of the studies have used cytosolic domains, as exemplified by the groundbreaking studies that elucidated the structure of a tetrapeptide helical bundle formed by interaction of the cytosolic domains of syntaxin1A, SNAP25 (two peptides) and synaptobrevin 2. Because the cytosolic fragments were unfettered by membrane attachments, it is likely that the tetrapeptide helical bundle reflects the lowest energy state, such as that found in the "cis" interactions of the SNARE motifs after fusion when they co-localize in the plasma membrane. Much more difficult to study and still poorly understood are critical "trans" interactions between the synaptic vesicle SNARE protein synaptobrevin 2 and the plasma membrane syntaxin1A/SNAP25 complex that initiate the fusion event. In a series of articles from the laboratory of Lukas Tamm, the spontaneous orientation of the SNARE motif of membrane-bound, full-length syntaxin1A with respect to the membrane hosting syntaxin's transmembrane domain was investigated with nanometer precision under a variety of conditions, including those that model aspects of the "trans" configuration. The studies rely on fluorescence interference-contrast microscopy, a technique that utilizes the pattern of constructive and destructive interference arising from incoming and reflected excitation and emission light at the surface of a silicon chip that has been layered with oxidized silicon of varying depths. This Perspective discusses their findings, including the unexpected influence of the degree of lipid unsaturation on the orientation of the SNARE complex.
Collapse
Affiliation(s)
- Ronald W Holz
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan.
| | - Joshua Zimmerberg
- Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
22
|
Shaaban A, Dhara M, Frisch W, Harb A, Shaib AH, Becherer U, Bruns D, Mohrmann R. The SNAP-25 linker supports fusion intermediates by local lipid interactions. eLife 2019; 8:41720. [PMID: 30883328 PMCID: PMC6422494 DOI: 10.7554/elife.41720] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/05/2019] [Indexed: 12/22/2022] Open
Abstract
SNAP-25 is an essential component of SNARE complexes driving fast Ca2+-dependent exocytosis. Yet, the functional implications of the tandem-like structure of SNAP-25 are unclear. Here, we have investigated the mechanistic role of the acylated “linker” domain that concatenates the two SNARE motifs within SNAP-25. Refuting older concepts of an inert connector, our detailed structure-function analysis in murine chromaffin cells demonstrates that linker motifs play a crucial role in vesicle priming, triggering, and fusion pore expansion. Mechanistically, we identify two synergistic functions of the SNAP-25 linker: First, linker motifs support t-SNARE interactions and accelerate ternary complex assembly. Second, the acylated N-terminal linker segment engages in local lipid interactions that facilitate fusion triggering and pore evolution, putatively establishing a favorable membrane configuration by shielding phospholipid headgroups and affecting curvature. Hence, the linker is a functional part of the fusion complex that promotes secretion by SNARE interactions as well as concerted lipid interplay.
Collapse
Affiliation(s)
- Ahmed Shaaban
- ZHMB, Saarland University, Homburg, Germany.,Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Madhurima Dhara
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Walentina Frisch
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Ali Harb
- ZHMB, Saarland University, Homburg, Germany
| | - Ali H Shaib
- Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Ute Becherer
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Dieter Bruns
- Institute for Physiology, Center of Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Ralf Mohrmann
- ZHMB, Saarland University, Homburg, Germany.,Institute for Physiology, Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Science, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
23
|
Chanaday NL, Kavalali ET. Time course and temperature dependence of synaptic vesicle endocytosis. FEBS Lett 2018; 592:3606-3614. [DOI: 10.1002/1873-3468.13268] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/02/2018] [Accepted: 10/06/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Natali L. Chanaday
- Department of Pharmacology; Vanderbilt Brain Institute; Vanderbilt University; Nashville TN USA
| | - Ege T. Kavalali
- Department of Pharmacology; Vanderbilt Brain Institute; Vanderbilt University; Nashville TN USA
| |
Collapse
|
24
|
Somasundaram A, Taraska JW. Local protein dynamics during microvesicle exocytosis in neuroendocrine cells. Mol Biol Cell 2018; 29:1891-1903. [PMID: 29874123 PMCID: PMC6085826 DOI: 10.1091/mbc.e17-12-0716] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium-triggered exocytosis is key to many physiological processes, including neurotransmitter and hormone release by neurons and endocrine cells. Dozens of proteins regulate exocytosis, yet the temporal and spatial dynamics of these factors during vesicle fusion remain unclear. Here we use total internal reflection fluorescence microscopy to visualize local protein dynamics at single sites of exocytosis of small synaptic-like microvesicles in live cultured neuroendocrine PC12 cells. We employ two-color imaging to simultaneously observe membrane fusion (using vesicular acetylcholine ACh transporter tagged to pHluorin) and the dynamics of associated proteins at the moments surrounding exocytosis. Our experiments show that many proteins, including the SNAREs syntaxin1 and VAMP2, the SNARE modulator tomosyn, and Rab proteins, are preclustered at fusion sites and rapidly lost at fusion. The ATPase N-ethylmaleimide–sensitive factor is locally recruited at fusion. Interestingly, the endocytic Bin-Amphiphysin-Rvs domain–containing proteins amphiphysin1, syndapin2, and endophilins are dynamically recruited to fusion sites and slow the loss of vesicle membrane-bound cargo from fusion sites. A similar effect on vesicle membrane protein dynamics was seen with the overexpression of the GTPases dynamin1 and dynamin2. These results suggest that proteins involved in classical clathrin-mediated endocytosis can regulate exocytosis of synaptic-like microvesicles. Our findings provide insights into the dynamics, assembly, and mechanistic roles of many key factors of exocytosis and endocytosis at single sites of microvesicle fusion in live cells.
Collapse
Affiliation(s)
- Agila Somasundaram
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
25
|
Sahu A, Sauer RA, Mandadapu KK. Irreversible thermodynamics of curved lipid membranes. Phys Rev E 2017; 96:042409. [PMID: 29347561 DOI: 10.1103/physreve.96.042409] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Indexed: 01/15/2023]
Abstract
The theory of irreversible thermodynamics for arbitrarily curved lipid membranes is presented here. The coupling between elastic bending and irreversible processes such as intramembrane lipid flow, intramembrane phase transitions, and protein binding and diffusion is studied. The forms of the entropy production for the irreversible processes are obtained, and the corresponding thermodynamic forces and fluxes are identified. Employing the linear irreversible thermodynamic framework, the governing equations of motion along with appropriate boundary conditions are provided.
Collapse
Affiliation(s)
- Amaresh Sahu
- Department of Chemical & Biomolecular Engineering, University of California at Berkeley, Berkeley, California 94720, USA
| | - Roger A Sauer
- Aachen Institute for Advanced Study in Computational Engineering Science (AICES), RWTH Aachen University, Templergraben 55, 52056 Aachen, Germany
| | - Kranthi K Mandadapu
- Department of Chemical & Biomolecular Engineering, University of California at Berkeley, Berkeley, California 94720, USA
| |
Collapse
|
26
|
Dhara M, Mohrmann R, Bruns D. v-SNARE function in chromaffin cells. Pflugers Arch 2017; 470:169-180. [PMID: 28887593 PMCID: PMC5748422 DOI: 10.1007/s00424-017-2066-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 01/04/2023]
Abstract
Vesicle fusion is elementary for intracellular trafficking and release of signal molecules, thus providing the basis for diverse forms of intercellular communication like hormonal regulation or synaptic transmission. A detailed characterization of the mechanisms underlying exocytosis is key to understand how the nervous system integrates information and generates appropriate responses to stimuli. The machinery for vesicular release employs common molecular players in different model systems including neuronal and neuroendocrine cells, in particular members of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) protein family, Sec1/Munc18-like proteins, and other accessory factors. To achieve temporal precision and speed, excitable cells utilize specialized regulatory proteins like synaptotagmin and complexin, whose interplay putatively synchronizes vesicle fusion and enhances stimulus-secretion coupling. In this review, we aim to highlight recent progress and emerging views on the molecular mechanisms, by which constitutively forming SNAREpins are organized in functional, tightly regulated units for synchronized release. Specifically, we will focus on the role of vesicle associated membrane proteins, also referred to as vesicular SNAREs, in fusion and rapid cargo discharge. We will further discuss the functions of SNARE regulators during exocytosis and focus on chromaffin cell as a model system of choice that allows for detailed structure-function analyses and direct measurements of vesicle fusion under precise control of intracellular [Ca]i.
Collapse
Affiliation(s)
- Madhurima Dhara
- Molecular Neurophysiology, CIPMM, Medical Faculty, Saarland University, 66421, Homburg/Saar, Germany
| | - Ralf Mohrmann
- Zentrum für Human- und Molekularbiologie, Saarland University, 66421, Homburg/Saar, Germany
| | - Dieter Bruns
- Molecular Neurophysiology, CIPMM, Medical Faculty, Saarland University, 66421, Homburg/Saar, Germany.
| |
Collapse
|
27
|
Abstract
Exocytosis is an important cellular process controlled by metabolic signaling. It involves vesicle fusion to the plasma membrane, followed by the opening of a fusion pore, and the subsequent release of the vesicular lumen content into the extracellular space. While most modeling efforts focus on the events leading to membrane fusion, how the vesicular membrane remodels after fusing to plasma membrane remains unclear. This latter event dictates the nature and the efficiency of exocytotic vesicular secretions, and is thus critical for exocytotic function. We provide a generic membrane mechanical model to systematically study the fate of post-fusion vesicles. We show that while membrane stiffness favors full-collapse vesicle fusion into the plasma membrane, the intravesicular pressure swells the vesicle and causes the fusion pore to shrink. Dimensions of the vesicle and its associated fusion pore further modulate this mechanical antagonism. We systematically define the mechanical conditions that account for the full spectrum of the observed vesicular secretion modes. Our model therefore can serve as a unified theoretical framework that sheds light on the elaborate control mechanism of exocytosis.
Collapse
Affiliation(s)
- Thomas Stephens
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States of America. Equal contribution
| | | | | |
Collapse
|
28
|
Chang CW, Chiang CW, Jackson MB. Fusion pores and their control of neurotransmitter and hormone release. J Gen Physiol 2017; 149:301-322. [PMID: 28167663 PMCID: PMC5339513 DOI: 10.1085/jgp.201611724] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/06/2017] [Accepted: 01/19/2017] [Indexed: 11/20/2022] Open
Abstract
Chang et al. review fusion pore structure and dynamics and discuss the implications for hormone and neurotransmitter release Ca2+-triggered exocytosis functions broadly in the secretion of chemical signals, enabling neurons to release neurotransmitters and endocrine cells to release hormones. The biological demands on this process can vary enormously. Although synapses often release neurotransmitter in a small fraction of a millisecond, hormone release can be orders of magnitude slower. Vesicles usually contain multiple signaling molecules that can be released selectively and conditionally. Cells are able to control the speed, concentration profile, and content selectivity of release by tuning and tailoring exocytosis to meet different biological demands. Much of this regulation depends on the fusion pore—the aqueous pathway by which molecules leave a vesicle and move out into the surrounding extracellular space. Studies of fusion pores have illuminated how cells regulate secretion. Furthermore, the formation and growth of fusion pores serve as a readout for the progress of exocytosis, thus revealing key kinetic stages that provide clues about the underlying mechanisms. Herein, we review the structure, composition, and dynamics of fusion pores and discuss the implications for molecular mechanisms as well as for the cellular regulation of neurotransmitter and hormone release.
Collapse
Affiliation(s)
- Che-Wei Chang
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705
| | - Chung-Wei Chiang
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705
| | - Meyer B Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
29
|
Cholesterol Increases the Openness of SNARE-Mediated Flickering Fusion Pores. Biophys J 2016; 110:1538-1550. [PMID: 27074679 DOI: 10.1016/j.bpj.2016.02.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 11/22/2022] Open
Abstract
Flickering of fusion pores during exocytotic release of hormones and neurotransmitters is well documented, but without assays that use biochemically defined components and measure single-pore dynamics, the mechanisms remain poorly understood. We used total internal reflection fluorescence microscopy to quantify fusion-pore dynamics in vitro and to separate the roles of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and lipid bilayer properties. When small unilamellar vesicles bearing neuronal v-SNAREs fused with planar bilayers reconstituted with cognate t-SNARES, lipid and soluble cargo transfer rates were severely reduced, suggesting that pores flickered. From the lipid release times we computed pore openness, the fraction of time the pore is open, which increased dramatically with cholesterol. For most lipid compositions tested, SNARE-mediated and nonspecifically nucleated pores had similar openness, suggesting that pore flickering was controlled by lipid bilayer properties. However, with physiological cholesterol levels, SNAREs substantially increased the fraction of fully open pores and fusion was so accelerated that there was insufficient time to recruit t-SNAREs to the fusion site, consistent with t-SNAREs being preclustered by cholesterol into functional docking and fusion platforms. Our results suggest that cholesterol opens pores directly by reducing the fusion-pore bending energy, and indirectly by concentrating several SNAREs into individual fusion events.
Collapse
|
30
|
Dhara M, Yarzagaray A, Makke M, Schindeldecker B, Schwarz Y, Shaaban A, Sharma S, Böckmann RA, Lindau M, Mohrmann R, Bruns D. v-SNARE transmembrane domains function as catalysts for vesicle fusion. eLife 2016; 5:e17571. [PMID: 27343350 PMCID: PMC4972536 DOI: 10.7554/elife.17571] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/24/2016] [Indexed: 12/22/2022] Open
Abstract
Vesicle fusion is mediated by an assembly of SNARE proteins between opposing membranes, but it is unknown whether transmembrane domains (TMDs) of SNARE proteins serve mechanistic functions that go beyond passive anchoring of the force-generating SNAREpin to the fusing membranes. Here, we show that conformational flexibility of synaptobrevin-2 TMD is essential for efficient Ca(2+)-triggered exocytosis and actively promotes membrane fusion as well as fusion pore expansion. Specifically, the introduction of helix-stabilizing leucine residues within the TMD region spanning the vesicle's outer leaflet strongly impairs exocytosis and decelerates fusion pore dilation. In contrast, increasing the number of helix-destabilizing, ß-branched valine or isoleucine residues within the TMD restores normal secretion but accelerates fusion pore expansion beyond the rate found for the wildtype protein. These observations provide evidence that the synaptobrevin-2 TMD catalyzes the fusion process by its structural flexibility, actively setting the pace of fusion pore expansion.
Collapse
Affiliation(s)
- Madhurima Dhara
- Institute for Physiology, Saarland University, Homburg, Germany
| | | | - Mazen Makke
- Institute for Physiology, Saarland University, Homburg, Germany
| | | | - Yvonne Schwarz
- Institute for Physiology, Saarland University, Homburg, Germany
| | - Ahmed Shaaban
- Zentrum für Human- und Molekularbiologie, Saarland University, Homburg, Germany
| | - Satyan Sharma
- Group Nanoscale Cell Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Rainer A Böckmann
- Computational Biology, Department of Biology, Friedrich-Alexander University, Erlangen, Germany
| | - Manfred Lindau
- Group Nanoscale Cell Biology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ralf Mohrmann
- Zentrum für Human- und Molekularbiologie, Saarland University, Homburg, Germany
| | - Dieter Bruns
- Institute for Physiology, Saarland University, Homburg, Germany
| |
Collapse
|
31
|
Trexler AJ, Sochacki KA, Taraska JW. Imaging the recruitment and loss of proteins and lipids at single sites of calcium-triggered exocytosis. Mol Biol Cell 2016; 27:2423-34. [PMID: 27307587 PMCID: PMC4966983 DOI: 10.1091/mbc.e16-01-0057] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/09/2016] [Indexed: 12/13/2022] Open
Abstract
Imaging of exocytic and endocytic proteins shows which are present at exocytic sites before, during, and after exocytosis in living cells. Rab proteins and SNARE modulators are lost, and dynamin, PIP2, and BAR-domain proteins are rapidly and transiently recruited, where they may modulate the nascent fusion pore. How and when the dozens of molecules that control exocytosis assemble in living cells to regulate the fusion of a vesicle with the plasma membrane is unknown. Here we image with two-color total internal reflection fluorescence microscopy the local changes of 27 proteins at single dense-core vesicles undergoing calcium-triggered fusion. We identify two broad dynamic behaviors of exocytic molecules. First, proteins enriched at exocytic sites are associated with DCVs long before exocytosis, and near the time of membrane fusion, they diffuse away. These proteins include Rab3 and Rab27, rabphilin3a, munc18a, tomosyn, and CAPS. Second, we observe a group of classical endocytic proteins and lipids, including dynamins, amphiphysin, syndapin, endophilin, and PIP2, which are rapidly and transiently recruited to the exocytic site near the time of membrane fusion. Dynamin mutants unable to bind amphiphysin were not recruited, indicating that amphiphysin is involved in localizing dynamin to the fusion site. Expression of mutant dynamins and knockdown of endogenous dynamin altered the rate of cargo release from single vesicles. Our data reveal the dynamics of many key proteins involved in exocytosis and identify a rapidly recruited dynamin/PIP2/BAR assembly that regulates the exocytic fusion pore of dense-core vesicles in cultured endocrine beta cells.
Collapse
Affiliation(s)
- Adam J Trexler
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kem A Sochacki
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
32
|
Kuchipudi A, Arroyo-Olarte RD, Hoffmann F, Brinkmann V, Gupta N. Optogenetic monitoring identifies phosphatidylthreonine-regulated calcium homeostasis in Toxoplasma gondii. MICROBIAL CELL 2016; 3:215-223. [PMID: 28357357 PMCID: PMC5349149 DOI: 10.15698/mic2016.05.500] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite, which inflicts acute as well as chronic infections in a wide range of warm-blooded vertebrates. Our recent work has demonstrated the natural occurrence and autonomous synthesis of an exclusive lipid phosphatidylthreonine in T. gondii. Targeted gene disruption of phosphatidylthreonine synthase impairs the parasite virulence due to unforeseen attenuation of the consecutive events of motility, egress and invasion. However, the underlying basis of such an intriguing phenotype in the parasite mutant remains unknown. Using an optogenetic sensor (gene-encoded calcium indicator, GCaMP6s), we show that loss of phosphatidylthreonine depletes calcium stores in intracellular tachyzoites, which leads to dysregulation of calcium release into the cytosol during the egress phase of the mutant. Consistently, the parasite motility and egress phenotypes in the mutant can be entirely restored by ionophore-induced mobilization of calcium. Collectively, our results suggest a novel regulatory function of phosphatidylthreonine in calcium signaling of a prevalent parasitic protist. Moreover, our application of an optogenetic sensor to monitor subcellular calcium in a model intracellular pathogen exemplifies its wider utility to other entwined systems.
Collapse
Affiliation(s)
| | | | | | | | - Nishith Gupta
- Humboldt University, Berlin, Germany.,Max-Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
33
|
Chang CW, Jackson MB. Synaptobrevin transmembrane domain influences exocytosis by perturbing vesicle membrane curvature. Biophys J 2016; 109:76-84. [PMID: 26153704 DOI: 10.1016/j.bpj.2015.05.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/29/2015] [Accepted: 05/11/2015] [Indexed: 01/12/2023] Open
Abstract
Membrane fusion requires that nearly flat lipid bilayers deform into shapes with very high curvature. This makes membrane bending a critical force in determining fusion mechanisms. A lipid bilayer will bend spontaneously when material is distributed asymmetrically between its two monolayers, and its spontaneous curvature (C0) will influence the stability of curved fusion intermediates. Prior work on Ca(2+)-triggered exocytosis revealed that fusion pore lifetime (τ) varies with vesicle content (Q), and showed that this relation reflects membrane bending energetics. Lipids that alter C0 change the dependence of τ on Q. These results suggested that the greater stability of an initial exocytotic fusion pore associated with larger vesicles reflects the need to bend more membrane during fusion pore dilation. In this study, we explored the possibility of manipulating C0 by mutating the transmembrane domain (TMD) of the vesicle membrane protein synaptobrevin 2 (syb2). Amperometric measurements of exocytosis in mouse chromaffin cells revealed that syb2 TMD mutations altered the relation between τ and Q. The effects of these mutations showed a striking periodicity, changing sign as the structural perturbation moved through the inner and outer leaflets. Some glycine and charge mutations also influenced the dependence of τ on Q in a manner consistent with expected changes in C0. These results suggest that side chains in the syb2 TMD influence the kinetics of exocytosis by perturbing the packing of the surrounding lipids. The present results support the view that membrane bending occurs during fusion pore expansion rather than during fusion pore formation. This supports the view of an initial fusion pore through two relatively flat membranes formed by protein.
Collapse
Affiliation(s)
- Che-Wei Chang
- Department of Neuroscience, Physiology Graduate Training Program, University of Wisconsin, Madison, Wisconsin
| | - Meyer B Jackson
- Department of Neuroscience, Physiology Graduate Training Program, University of Wisconsin, Madison, Wisconsin.
| |
Collapse
|
34
|
Zelkas L, Raghupathi R, Lumsden AL, Martin AM, Sun E, Spencer NJ, Young RL, Keating DJ. Serotonin-secreting enteroendocrine cells respond via diverse mechanisms to acute and chronic changes in glucose availability. Nutr Metab (Lond) 2015; 12:55. [PMID: 26673561 PMCID: PMC4678665 DOI: 10.1186/s12986-015-0051-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/09/2015] [Indexed: 01/11/2023] Open
Abstract
Background Enteroendocrine cells collectively constitute our largest endocrine tissue, with serotonin (5-HT) secreting enterochromaffin (EC) cells being the largest component (~50 %). This gut-derived 5-HT has multiple paracrine and endocrine roles. EC cells are thought to act as nutrient sensors and luminal glucose is the major absorbed form of carbohydrate in the gut and activates secretion in an array of cell types. It is unknown whether EC cells release 5-HT in response to glucose in primary EC cells. Furthermore, fasting augments 5-HT synthesis and release into the circulation. However, which nutrients cause fasting-induced synthesis of EC cell 5-HT is unknown. Here we examine the effects of acute and chronic changes in glucose availability on 5-HT release from intact tissue and single EC cells. Methods We utilised established approaches in our laboratories measuring 5-HT release in intact mouse colon with amperometry. We then examined single EC cells function using our published protocol in guinea-pig colon. Single cell Ca2+ imaging and amperometry were used with these cells. Real-time PCR was used along with amperometry, on primary EC cells cultured for 24 h in 5 or 25 mM glucose. Results We demonstrate that acute increases in glucose, at levels found in the gut lumen rather than in plasma, trigger 5-HT release from intact colon, and cause Ca2+ entry and 5-HT release in primary EC cells. Single cell amperometry demonstrates that high glucose increases the amount of 5-HT released from individual vesicles as they undergo exocytosis. Finally, 24 h incubation of EC cells in low glucose causes an increase in the transcription of the 5-HT synthesising enzyme Tph1 as well as increasing in 5-HT secretion in EC cells. Conclusions We demonstrate that primary EC cells respond to acute changes in glucose availability through increases in intracellular Ca2+ the activation of 5-HT secretion, but respond to chronic changes in glucose levels through the transcriptional regulation of Tph1 to alter 5-HT synthesis.
Collapse
Affiliation(s)
- Leah Zelkas
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia
| | - Ravi Raghupathi
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia ; South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 Australia
| | - Amanda L Lumsden
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia ; South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 Australia
| | - Alyce M Martin
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia ; South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 Australia
| | - Emily Sun
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia ; South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 Australia
| | - Nick J Spencer
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia
| | - Richard L Young
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 Australia ; Discipline of Medicine, University of Adelaide, Adelaide, SA 5001 Australia
| | - Damien J Keating
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Rd, Adelaide, SA 5042 Australia ; South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 Australia
| |
Collapse
|
35
|
Chang CW, Chiang CW, Gaffaney JD, Chapman ER, Jackson MB. Lipid-anchored Synaptobrevin Provides Little or No Support for Exocytosis or Liposome Fusion. J Biol Chem 2015; 291:2848-57. [PMID: 26663078 DOI: 10.1074/jbc.m115.701169] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Indexed: 12/11/2022] Open
Abstract
SNARE proteins catalyze many forms of biological membrane fusion, including Ca(2+)-triggered exocytosis. Although fusion mediated by SNAREs generally involves proteins anchored to each fusing membrane by a transmembrane domain (TMD), the role of TMDs remains unclear, and previous studies diverge on whether SNAREs can drive fusion without a TMD. This issue is important because it relates to the question of the structure and composition of the initial fusion pore, as well as the question of whether SNAREs mediate fusion solely by creating close proximity between two membranes versus a more active role in transmitting force to the membrane to deform and reorganize lipid bilayer structure. To test the role of membrane attachment, we generated four variants of the synaptic v-SNARE synaptobrevin-2 (syb2) anchored to the membrane by lipid instead of protein. These constructs were tested for functional efficacy in three different systems as follows: Ca(2+)-triggered dense core vesicle exocytosis, spontaneous synaptic vesicle exocytosis, and Ca(2+)-synaptotagmin-enhanced SNARE-mediated liposome fusion. Lipid-anchoring motifs harboring one or two lipid acylation sites completely failed to support fusion in any of these assays. Only the lipid-anchoring motif from cysteine string protein-α, which harbors many lipid acylation sites, provided support for fusion but at levels well below that achieved with wild type syb2. Thus, lipid-anchored syb2 provides little or no support for exocytosis, and anchoring syb2 to a membrane by a TMD greatly improves its function. The low activity seen with syb2-cysteine string protein-α may reflect a slower alternative mode of SNARE-mediated membrane fusion.
Collapse
Affiliation(s)
| | | | | | - Edwin R Chapman
- Biophysics Ph.D. Program, Howard Hughes Medical Institute, and Department of Neuroscience, University of Wisconsin, Madison, Wisconsin 53705
| | - Meyer B Jackson
- From the Physiology Ph.D. Graduate Training Program, Biophysics Ph.D. Program, Department of Neuroscience, University of Wisconsin, Madison, Wisconsin 53705
| |
Collapse
|
36
|
Arroyo-Olarte RD, Brouwers JF, Kuchipudi A, Helms JB, Biswas A, Dunay IR, Lucius R, Gupta N. Phosphatidylthreonine and Lipid-Mediated Control of Parasite Virulence. PLoS Biol 2015; 13:e1002288. [PMID: 26565995 PMCID: PMC4643901 DOI: 10.1371/journal.pbio.1002288] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 09/30/2015] [Indexed: 01/10/2023] Open
Abstract
The major membrane phospholipid classes, described thus far, include phosphatidylcholine (PtdCho), phosphatidylethanolamine (PtdEtn), phosphatidylserine (PtdSer), and phosphatidylinositol (PtdIns). Here, we demonstrate the natural occurrence and genetic origin of an exclusive and rather abundant lipid, phosphatidylthreonine (PtdThr), in a common eukaryotic model parasite, Toxoplasma gondii. The parasite expresses a novel enzyme PtdThr synthase (TgPTS) to produce this lipid in its endoplasmic reticulum. Genetic disruption of TgPTS abrogates de novo synthesis of PtdThr and impairs the lytic cycle and virulence of T. gondii. The observed phenotype is caused by a reduced gliding motility, which blights the parasite egress and ensuing host cell invasion. Notably, the PTS mutant can prevent acute as well as yet-incurable chronic toxoplasmosis in a mouse model, which endorses its potential clinical utility as a metabolically attenuated vaccine. Together, the work also illustrates the functional speciation of two evolutionarily related membrane phospholipids, i.e., PtdThr and PtdSer. An exclusive membrane lipid, phosphatidylthreonine, is revealed to be naturally abundant in the widespread protist parasite Toxoplasma gondii, where it has evolved adaptively and is essential for parasite virulence. Lipids are essential constituents of biological membranes, and most organisms across the tree of life use a relatively limited repertoire of lipids in their membranes. This work reveals the natural and abundant presence of an exclusive lipid phosphatidylthreonine (PtdThr) in Toxoplasma gondii, a ubiquitous protozoan parasite of humans and animals. PtdThr is made by a novel parasite-specific enzyme, PtdThr synthase, which has evolved from the widespread enzyme phosphatidylserine synthase. The study shows that PtdThr is required for asexual reproduction and virulence of the parasite in vivo, and a metabolically attenuated mutant strain of Toxoplasma lacking PtdThr can protect vaccinated mice against acute and currently incurable chronic infection. This discovery demonstrates adaptive “speciation” of PtdThr from an otherwise near-universal membrane lipid phosphatidylserine and reveals de novo PtdThr synthesis in T. gondii as a potential drug target.
Collapse
Affiliation(s)
| | - Jos F. Brouwers
- Department of Biochemistry and Cell Biology, Institute of Biomembranes, Utrecht University, The Netherlands
| | - Arunakar Kuchipudi
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
| | - J. Bernd Helms
- Department of Biochemistry and Cell Biology, Institute of Biomembranes, Utrecht University, The Netherlands
| | - Aindrila Biswas
- Institute of Medical Microbiology, Otto von Guericke University, Magdeburg, Germany
| | - Ildiko R. Dunay
- Institute of Medical Microbiology, Otto von Guericke University, Magdeburg, Germany
| | - Richard Lucius
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
| | - Nishith Gupta
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
- Parasitology Unit, Max-Planck Institute for Infection Biology, Berlin, Germany
- * E-mail:
| |
Collapse
|
37
|
Chang CW, Hui E, Bai J, Bruns D, Chapman ER, Jackson MB. A structural role for the synaptobrevin 2 transmembrane domain in dense-core vesicle fusion pores. J Neurosci 2015; 35:5772-80. [PMID: 25855187 PMCID: PMC4388931 DOI: 10.1523/jneurosci.3983-14.2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 03/01/2015] [Accepted: 03/03/2015] [Indexed: 01/17/2023] Open
Abstract
Ca(2+)-triggered release of neurotransmitters and hormones depends on soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) to drive the fusion of the vesicle and plasma membranes. The formation of the SNARE complex by the vesicle SNARE synaptobrevin 2 (syb2) and the two plasma membrane SNAREs syntaxin (syx) and SNAP-25 draws the two membranes together, but the events that follow membrane juxtaposition, and the ways that SNAREs remodel lipid membranes remain poorly understood. The SNAREs syx and syb2 have transmembrane domains (TMDs) that can exert force directly on the lipid bilayers. The TMD of syx influences fusion pore flux in a manner that suggests it lines the nascent fusion pore through the plasma membrane. The TMD of syb2 traverses the vesicle membrane and is the most likely partner to syx in completing a proteinaceous fusion pore through the vesicle membrane, but the role of this vesicle SNARE in fusion pores has yet to be tested. Here amperometry and conductance measurements were performed to probe the function of the syb2 TMD in fusion pores formed during catecholamine exocytosis in mouse chromaffin cells. Fusion pore flux was sensitive to the size and charge of TMD residues near the N terminus; fusion pore conductance was altered by substitutions at these sites. Unlike syx, the syb2 residues that influence fusion pore permeation fell along two α-helical faces of its TMD, rather than one. These results indicate a role for the syb2 TMD in nascent fusion pores, but in a very different structural arrangement from that of the syx TMD.
Collapse
Affiliation(s)
- Che-Wei Chang
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin 53705
| | - Enfu Hui
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, California 94158
| | - Jihong Bai
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Dieter Bruns
- Institut für Physiologie, Universität des Saarlandes, 66424 Homburg, Germany, and
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin 53705, Howard Hughes Medical Institute, University of Wisconsin, Madison, Wisconsin 53706
| | - Meyer B Jackson
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin 53705,
| |
Collapse
|
38
|
Koseoglu S, Meyer A, Kim D, Meyer BM, Wang Y, Dalluge JJ, Haynes CL. Analytical characterization of the role of phospholipids in platelet adhesion and secretion. Anal Chem 2015; 87:413-21. [PMID: 25439269 PMCID: PMC4287828 DOI: 10.1021/ac502293p] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 12/02/2014] [Indexed: 12/19/2022]
Abstract
The cellular phospholipid membrane plays an important role in cell function and cell-cell communication, but its biocomplexity and dynamic nature presents a challenge for examining cellular uptake of phospholipids and the resultant effects on cell function. Platelets, small anuclear circulating cell bodies that influence a wide variety of physiological functions through their dynamic secretory and adhesion behavior, present an ideal platform for exploring the effects of exogenous phospholipids on membrane phospholipid content and cell function. In this work, a broad range of platelet functions are quantitatively assessed by leveraging a variety of analytical chemistry techniques, including ultraperformance liquid chromatography-tandem electrospray ionization mass spectrometry (UPLC-MS/MS), vasculature-mimicking microfluidic analysis, and single cell carbon-fiber microelectrode amperometry (CFMA). The relative enrichments of phosphatidylserine (PS) and phosphatidylethanolamine (PE) were characterized with UPLC-MS/MS, and the effects of the enrichment of these two phospholipids on both platelet secretory behavior and adhesion were examined. Results show that, in fact, both PS and PE influence platelet adhesion and secretion. PS was enriched dramatically and decreased platelet adhesion as well as secretion from δ-, α-, and lysosomal granules. PE enrichment was moderate and increased secretion from platelet lysosomes. These insights illuminate the critical connection between membrane phospholipid character and platelet behavior, and both the methods and results presented herein are likely translatable to other mammalian cell systems.
Collapse
Affiliation(s)
- Secil Koseoglu
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Audrey
F. Meyer
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Donghyuk Kim
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Ben M. Meyer
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Yiwen Wang
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Joseph J. Dalluge
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Christy L. Haynes
- University of Minnesota, Department of
Chemistry, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
39
|
Mackenzie KD, Duffield MD, Peiris H, Phillips L, Zanin MP, Teo EH, Zhou XF, Keating DJ. Huntingtin-associated protein 1 regulates exocytosis, vesicle docking, readily releasable pool size and fusion pore stability in mouse chromaffin cells. J Physiol 2013; 592:1505-18. [PMID: 24366265 DOI: 10.1113/jphysiol.2013.268342] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Huntingtin-associated protein 1 (HAP1) was initially established as a neuronal binding partner of huntingtin, mutations in which underlie Huntington's disease. Subcellular localization and protein interaction data indicate that HAP1 may be important in vesicle trafficking and cell signalling. In this study, we establish that HAP1 is important in several steps of exocytosis in adrenal chromaffin cells. Using carbon-fibre amperometry, we measured single vesicle exocytosis in chromaffin cells obtained from HAP1(-/-) and HAP1(+/+) littermate mice. Numbers of Ca(2+)-dependent and Ca(2+)-independent full fusion events in HAP1(-/-) cells are significantly decreased compared with those in HAP1(+/+) cells. We observed no change in the frequency of 'kiss-and-run' fusion events or in Ca(2+) entry. Whereas release per full fusion event is unchanged in HAP1(-/-) cells, early fusion pore duration is prolonged, as indicated by the increased duration of pre-spike foot signals. Kiss-and-run events have a shorter duration, indicating opposing roles for HAP1 in the stabilization of the fusion pore during full fusion and transient fusion, respectively. We use electron microscopy to demonstrate a reduction in the number of vesicles docked at the plasma membrane of HAP1(-/-) cells, where membrane capacitance measurements reveal the readily releasable pool of vesicles to be reduced in size. Our study therefore illustrates that HAP1 regulates exocytosis by influencing the morphological docking of vesicles at the plasma membrane, the ability of vesicles to be released rapidly upon stimulation, and the early stages of fusion pore formation.
Collapse
Affiliation(s)
- Kimberly D Mackenzie
- Department of Human Physiology, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Raghupathi R, Duffield MD, Zelkas L, Meedeniya A, Brookes SJH, Sia TC, Wattchow DA, Spencer NJ, Keating DJ. Identification of unique release kinetics of serotonin from guinea-pig and human enterochromaffin cells. J Physiol 2013; 591:5959-75. [PMID: 24099799 PMCID: PMC3872764 DOI: 10.1113/jphysiol.2013.259796] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 10/02/2013] [Indexed: 12/21/2022] Open
Abstract
The major source of serotonin (5-HT) in the body is the enterochromaffin (EC) cells lining the intestinal mucosa of the gastrointestinal tract. Despite the fact that EC cells synthesise ∼95% of total body 5-HT, and that this 5-HT has important paracrine and endocrine roles, no studies have investigated the mechanisms of 5-HT release from single primary EC cells. We have developed a rapid primary culture of guinea-pig and human EC cells, allowing analysis of single EC cell function using electrophysiology, electrochemistry, Ca(2+) imaging, immunocytochemistry and 3D modelling. Ca(2+) enters EC cells upon stimulation and triggers quantal 5-HT release via L-type Ca(2+) channels. Real time amperometric techniques reveal that EC cells release 5-HT at rest and this release increases upon stimulation. Surprisingly for an endocrine cell storing 5-HT in large dense core vesicles (LDCVs), EC cells release 70 times less 5-HT per fusion event than catecholamine released from similarly sized LDCVs in endocrine chromaffin cells, and the vesicle release kinetics instead resembles that observed in mammalian synapses. Furthermore, we measured EC cell density along the gastrointestinal tract to create three-dimensional (3D) simulations of 5-HT diffusion using the minimal number of variables required to understand the physiological relevance of single cell 5-HT release in the whole-tissue milieu. These models indicate that local 5-HT levels are likely to be maintained around the activation threshold for mucosal 5-HT receptors and that this is dependent upon stimulation and location within the gastrointestinal tract. This is the first study demonstrating single cell 5-HT release in primary EC cells. The mode of 5-HT release may represent a unique mode of exocytosis amongst endocrine cells and is functionally relevant to gastrointestinal sensory and motor function.
Collapse
Affiliation(s)
- Ravinarayan Raghupathi
- D. Keating: Department of Human Physiology and Centre for Neuroscience, Flinders University, Sturt Road, Adelaide, 5001, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zanin MP, Mackenzie KD, Peiris H, Pritchard MA, Keating DJ. RCAN1 regulates vesicle recycling and quantal release kinetics via effects on calcineurin activity. J Neurochem 2013; 124:290-9. [PMID: 23134420 DOI: 10.1111/jnc.12086] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 10/11/2012] [Accepted: 11/06/2012] [Indexed: 11/29/2022]
Abstract
We have previously shown that Regulator of Calcineurin 1 (RCAN1) regulates multiple stages of vesicle exocytosis. However, the mechanisms by which RCAN1 affects secretory vesicle exocytosis and quantal release kinetics remain unknown. Here, we use carbon fibre amperometry to detect exocytosis from chromaffin cells and identify these underlying mechanisms. We observe reduced exocytosis with repeated stimulations in chromaffin cells over-expressing RCAN1 (RCAN1(ox)), but not in wild-type (WT) cells, indicating a negative effect of RCAN1 on vesicle recycling and endocytosis. Acute exposure to calcineurin inhibitors, cyclosporine A and FK-506, replicates this effect in WT cells but has no additional effect in RCAN1(ox) cells. When we chronically expose WT cells to cyclosporine A and FK-506 we find that catecholamine release per vesicle and pre-spike foot (PSF) signal parameters are decreased, similar to that in RCAN1(ox) cells. Inhibiting calcineurin activity in RCAN1(ox) cells has no additional effect on the amount of catecholamine release per vesicle but further reduces PSF signal parameters. Although electron microscopy studies indicate these changes are not because of altered vesicle number or distribution in RCAN1(ox) cells, the smaller vesicle and dense core size we observe in RCAN1(ox) cells may underlie the reduced quantal release in these cells. Thus, our results indicate that RCAN1 most likely affects vesicle recycling and quantal release kinetics via the inhibition of calcineurin activity.
Collapse
Affiliation(s)
- Mark P Zanin
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | | | | | | | | |
Collapse
|
42
|
Stenovec M, Gonçalves PP, Zorec R. Peptide hormone release monitored from single vesicles in "membrane lawns" of differentiated male pituitary cells: SNAREs and fusion pore widening. Endocrinology 2013; 154:1235-46. [PMID: 23372020 DOI: 10.1210/en.2012-2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study we used live-cell immunocytochemistry and confocal microscopy to study the release from a single vesicle in a simplified system called membrane lawns. The lawns were prepared by exposing differentiated pituitary prolactin (PRL)-secreting cells to a hypoosmotic shear stress. The density of the immunolabeled ternary soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE) complexes that bind complexin was approximately 10 times lower than the PRL-positive, lawn-resident vesicles; this indicates that some but not all vesicles are associated with ternary SNARE complexes. However, lawn-resident PRL vesicles colocalized relatively well with particular SNARE proteins: synaptobrevin 2 (35%), syntaxin 1 (22%), and 25-kDa synaptosome associated protein (6%). To study vesicle discharge, we prepared lawn-resident vesicles, derived from atrial natriuretic peptide tagged with emerald fluorescent protein (ANP.emd)-transfected cells, which label vesicles. These maintained the structural passage to the exterior because approximately 40% of ANP.emd-loaded vesicles were labeled by extracellular PRL antibodies. Cargo release from the lawn-resident vesicles, monitored by the decline in the ANP.emd fluorescence intensity, was similar to that in intact cells. It is likely that SNARE proteins are required for calcium-dependent release from these vesicles. This is because the expression of the dominant-negative SNARE peptide, which interferes with SNARE complex formation, reduced the number of PRL-positive spots per cell (PRL antibodies placed extracellularly) significantly, from 58 ± 9 to 4 ± 2. In dominant-negative SNARE-treated cells, the PRL-positive area was reduced from 0.259 ± 0.013 to 0.123 ± 0.014 μm(2), which is consistent with a hindered vesicle luminal access for extracellular PRL antibodies. These results indicate that vesicle discharge is regulated by SNARE-mediated fusion pore widening.
Collapse
Affiliation(s)
- Matjaž Stenovec
- Celica Biomedical Center, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | | | | |
Collapse
|
43
|
Garewal M, Zhang L, Ren G. Optimized negative-staining protocol for examining lipid-protein interactions by electron microscopy. Methods Mol Biol 2013; 974:111-8. [PMID: 23404274 PMCID: PMC10546916 DOI: 10.1007/978-1-62703-275-9_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
A large number of proteins are capable of inserting themselves into lipids, and interacting with membranes, such as transmembrane proteins and apolipoproteins. Protein-lipid interactions have been identified as one of the keys in understanding biological processes, while the structure of proteins at the lipid-binding stage can provide evidence to help identify their roles and critical functions. However, structure determination of proteins at the lipid-binding stage is rather difficult, because conformational and compositional heterogeneities of the protein-lipid complexes are major barriers to unravel their structures using traditional methods, such as X-ray crystallography. Electron microscopy (EM) is an alternative approach to determine protein structure and has demonstrated a capability in visualizing lipid-protein interactions directly. Among various EM techniques, negative-staining (NS) is an easy, rapid, qualitative approach that is a well-established technique, frequently used in research laboratories. Conventional NS protocols, unfortunately, often generate artifacts with lipid-related proteins, such as the rouleau formation of lipoproteins. To overcome this artifact formation, Ren and his colleagues recently developed an optimized NS protocol that was validated by comparing images of lipoproteins from cryo-electron microscopy (cryo-EM). The optimized NS protocol could produce "near native-state" particle images and high contrast images of the protein in its lipid-binding state that is favorable for three-dimensional (3D) reconstruction by single-particle analysis and individual-particle electron tomography (IPET), suggesting this optimized protocol can be used widely to examine the structure of proteins at lipid-binding stage.
Collapse
Affiliation(s)
- Mark Garewal
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | | |
Collapse
|
44
|
Zhang L, Ren G. 108 Individual-particle electron tomography: a method for studying macromolecule dynamics. J Biomol Struct Dyn 2013. [DOI: 10.1080/07391102.2013.786350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
45
|
Fang Q, Zhao Y, Lindau M. Juxtamembrane tryptophans of synaptobrevin 2 control the process of membrane fusion. FEBS Lett 2012. [PMID: 23178715 DOI: 10.1016/j.febslet.2012.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Synaptobrevin 2 (Syb2), syntaxin (Sx1A), and SNAP-25, generate a force to induce fusion pore formation. The v-SNARE, Syb2, is anchored to the vesicle membrane by a single transmembrane domain. Here we show that 2 tryptophans (W89/W90) located in the juxtamembrane domain of Syb2, which stabilize the transmembrane (TM) domain position, control the ratio of spontaneous vs. stimulated membrane fusion events in chromaffin cells. Changing the 2 hydrophobic tryptophans to neutral alanines promotes spontaneous membrane fusion, faster transmitter release kinetics and complete release from individual vesicles. The results indicate that the two tryptophans act as a fusion clamp making fusion stimulus-dependent.
Collapse
Affiliation(s)
- Qinghua Fang
- School of Applied & Engineering Physics, Cornell University, Ithaca, NY 14853, USA.
| | | | | |
Collapse
|
46
|
Exocytosis is impaired in mucopolysaccharidosis IIIA mouse chromaffin cells. Neuroscience 2012; 227:110-8. [PMID: 23022219 DOI: 10.1016/j.neuroscience.2012.09.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 08/28/2012] [Accepted: 09/13/2012] [Indexed: 11/22/2022]
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disorder caused by a deficiency in the activity of the lysosomal hydrolase, sulphamidase, an enzyme involved in the degradation of heparan sulphate. MPS IIIA patients exhibit progressive mental retardation and behavioural disturbance. While neuropathology is the major clinical problem in MPS IIIA patients, there is little understanding of how lysosomal storage generates this phenotype. As reduced neuronal communication can underlie cognitive deficiencies, we investigated whether the secretion of neurotransmitters is altered in MPS IIIA mice; utilising adrenal chromaffin cells, a classical model for studying secretion via exocytosis. MPS IIIA chromaffin cells displayed heparan sulphate storage and electron microscopy revealed large electron-lucent storage compartments. There were also increased numbers of large/elongated chromaffin granules, with a morphology that was similar to immature secretory granules. Carbon fibre amperometry illustrated a significant decrease in the number of exocytotic events for MPS IIIA, when compared to control chromaffin cells. However, there were no changes in the kinetics of release, the amount of catecholamine released per exocytotic event, or the amount of Ca(2+) entry upon stimulation. The increased number of large/elongated granules and reduced number of exocytotic events suggests that either the biogenesis and/or the cell surface docking and fusion potential of these vesicles is impaired in MPS IIIA. If this also occurs in central nervous system neurons, the reduction in neurotransmitter release could help to explain the development of neuropathology in MPS IIIA.
Collapse
|
47
|
Multiple roles for the actin cytoskeleton during regulated exocytosis. Cell Mol Life Sci 2012; 70:2099-121. [PMID: 22986507 DOI: 10.1007/s00018-012-1156-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/28/2012] [Accepted: 08/30/2012] [Indexed: 01/01/2023]
Abstract
Regulated exocytosis is the main mechanism utilized by specialized secretory cells to deliver molecules to the cell surface by virtue of membranous containers (i.e., secretory vesicles). The process involves a series of highly coordinated and sequential steps, which include the biogenesis of the vesicles, their delivery to the cell periphery, their fusion with the plasma membrane, and the release of their content into the extracellular space. Each of these steps is regulated by the actin cytoskeleton. In this review, we summarize the current knowledge regarding the involvement of actin and its associated molecules during each of the exocytic steps in vertebrates, and suggest that the overall role of the actin cytoskeleton during regulated exocytosis is linked to the architecture and the physiology of the secretory cells under examination. Specifically, in neurons, neuroendocrine, endocrine, and hematopoietic cells, which contain small secretory vesicles that undergo rapid exocytosis (on the order of milliseconds), the actin cytoskeleton plays a role in pre-fusion events, where it acts primarily as a functional barrier and facilitates docking. In exocrine and other secretory cells, which contain large secretory vesicles that undergo slow exocytosis (seconds to minutes), the actin cytoskeleton plays a role in post-fusion events, where it regulates the dynamics of the fusion pore, facilitates the integration of the vesicles into the plasma membrane, provides structural support, and promotes the expulsion of large cargo molecules.
Collapse
|
48
|
Jesenek D, Perutková S, Kralj-Iglič V, Kralj S, Iglič A. Exocytotic fusion pore stability and topological defects in the membrane with orientational degree of ordering. Cell Calcium 2012; 52:277-82. [PMID: 22541648 DOI: 10.1016/j.ceca.2012.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 03/26/2012] [Accepted: 04/02/2012] [Indexed: 11/30/2022]
Abstract
Regulated exocytosis is a process that strongly depends on the formation and stability of the fusion pore. It was indicated experimentally and theoretically that narrow and highly curved fusion pore may be stabilized by accumulation of anisotropic membrane components possessing orientational ordering. On the other hand, narrow fusion pore may also undergo repetitive opening and closing, disruption in the so called kiss and run process or become completely opened in the process of full fusion of the vesicle with the membrane. In this paper we attempt to elucidate the subtle interplay between the stabilizing and destabilizing processes in the fusion neck. A possible physical mechanism which may lead to disruption of the stable fusion pore or complete fusion of the vesicle with the membrane is discussed. It is indicated that topologically driven defects of the in-plane orientational membrane ordering in the region of the fusion pore may disrupt the fusion. Alternatively, it may facilitate repetitive opening and closing of the fusion pore or induce full fusion of the vesicle with the target membrane.
Collapse
Affiliation(s)
- Dalija Jesenek
- Department of Condensed Matter Physics, J. Stefan Institute, SI-1000 Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
49
|
Mohrmann R, Sørensen JB. SNARE requirements en route to exocytosis: from many to few. J Mol Neurosci 2012; 48:387-94. [PMID: 22427188 DOI: 10.1007/s12031-012-9744-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/29/2012] [Indexed: 12/30/2022]
Abstract
Although it has been known for almost two decades that the ternary complex of N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) constitutes the functional unit driving membrane fusion, our knowledge about the dynamical arrangement and organization of SNARE proteins and their complexes before and during vesicle exocytosis is still limited. Here, we review recent progress in this expanding field with emphasis on the question of fusion complex stoichiometry, i.e., how many SNARE proteins and complexes are needed for the fusion of a vesicle with the plasma membrane.
Collapse
Affiliation(s)
- Ralf Mohrmann
- Department of Physiology, University of Saarland, Homburg, Germany.
| | | |
Collapse
|
50
|
Jackson MB. Inferring structures of kinetic intermediates in Ca(2+)-triggered exocytosis. CURRENT TOPICS IN MEMBRANES 2012; 68:185-208. [PMID: 21771500 DOI: 10.1016/b978-0-12-385891-7.00008-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Affiliation(s)
- Meyer B Jackson
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|