1
|
Pieretti JC, Horne TL, García-Villasante N, Seabra AB, Muntané J. Zinc-Based Nanoparticles, but Not Silicon-Based Nanoparticles, Accumulate in Mitochondria and Promote Cell Death in Liver Cancer Cells. Int J Nanomedicine 2024; 19:12409-12420. [PMID: 39606560 PMCID: PMC11600939 DOI: 10.2147/ijn.s474643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/28/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is the main hepatic primary malignancy. Patients with advanced HCC receiving the recommended therapies have a poor outcome. In different settings, nanotechnology has gained attraction as a potential alternative strategy for improving therapeutic effectiveness. Among several nanoparticles (NPs), inorganic NPs, such as zinc and silicon oxides (ZnO and SiO2), are mainly chosen as drug nanocarriers, as both present great adsorption properties and biocompatibility. Aim The objective is to identify the molecular mechanisms underlying the proapoptotic effects of ZnO and SiO2 NPs in differentiated hepatoblastoma cells (HepG2) and mesenchymal liver cancer cells (SNU449). Methods Dose-dependent induction of cell cytotoxicity by ZnO and SiO2 NPs (5 to 50 µg/mL) was determined in HepG2 and SNU449 cells. NPs intracellular localization was assessed using transmission electron microscopy (TEM). Cell death was determined by trypan blue staining and caspase-3 and -8 activities. Cell respiration was determined using MitroStress assay (Seahorse, Agilent). Results ZnO NPs, but not SiO2 NPs, reduced cell viability in HepG2 and SNU449. Interestingly, SNU449 appeared to be more susceptible than HepG2 to ZnO NPs (IC50 of 27.4 ± 1.4 µg/mL and 41.8 ± 0.4 µg/mL, respectively). SiO2 NPs tended to be localized in lysosomes in both cell lines, while ZnO NPs demonstrated a random distribution with a high presence in mitochondria and related structures. As expected, SiO2 NPs did not reduce cell survival and cell respiration, while ZnO NPs promoted cell death and decreased oxygen consumption rate. ZnO NPs mitochondrial accumulation was associated with increased apoptosis in HepG2, while necroapoptosis was mainly involved in ZnO-induced cell death in SNU449. Conclusion SiO2 demonstrated no cytotoxic profile against liver cancer cells. ZnO NPs demonstrated to accumulate in mitochondria impacting cell respiration and cell death in liver cancer cells. ZnO induced apoptosis and necroptosis in HepG2 and SNU449, respectively.
Collapse
Affiliation(s)
- Joana C Pieretti
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, Seville, Spain
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP, Brazil
| | - Thaissa L Horne
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, Seville, Spain
| | - Natalia García-Villasante
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, Seville, Spain
| | - Amedea B Seabra
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP, Brazil
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain
| |
Collapse
|
2
|
Abela L, Gianfrancesco L, Tagliatti E, Rossignoli G, Barwick K, Zourray C, Reid KM, Budinger D, Ng J, Counsell J, Simpson A, Pearson TS, Edvardson S, Elpeleg O, Brodsky FM, Lignani G, Barral S, Kurian MA. Neurodevelopmental and synaptic defects in DNAJC6 parkinsonism, amenable to gene therapy. Brain 2024; 147:2023-2037. [PMID: 38242634 PMCID: PMC11146427 DOI: 10.1093/brain/awae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/10/2023] [Accepted: 12/16/2023] [Indexed: 01/21/2024] Open
Abstract
DNAJC6 encodes auxilin, a co-chaperone protein involved in clathrin-mediated endocytosis (CME) at the presynaptic terminal. Biallelic mutations in DNAJC6 cause a complex, early-onset neurodegenerative disorder characterized by rapidly progressive parkinsonism-dystonia in childhood. The disease is commonly associated with additional neurodevelopmental, neurological and neuropsychiatric features. Currently, there are no disease-modifying treatments for this condition, resulting in significant morbidity and risk of premature mortality. To investigate the underlying disease mechanisms in childhood-onset DNAJC6 parkinsonism, we generated induced pluripotent stem cells (iPSC) from three patients harbouring pathogenic loss-of-function DNAJC6 mutations and subsequently developed a midbrain dopaminergic neuronal model of disease. When compared to age-matched and CRISPR-corrected isogenic controls, the neuronal cell model revealed disease-specific auxilin deficiency as well as disturbance of synaptic vesicle recycling and homeostasis. We also observed neurodevelopmental dysregulation affecting ventral midbrain patterning and neuronal maturation. To explore the feasibility of a viral vector-mediated gene therapy approach, iPSC-derived neuronal cultures were treated with lentiviral DNAJC6 gene transfer, which restored auxilin expression and rescued CME. Our patient-derived neuronal model provides deeper insights into the molecular mechanisms of auxilin deficiency as well as a robust platform for the development of targeted precision therapy approaches.
Collapse
Affiliation(s)
- Lucia Abela
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Lorita Gianfrancesco
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Erica Tagliatti
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, 20089 Milano, Italy
| | - Giada Rossignoli
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Katy Barwick
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Clara Zourray
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kimberley M Reid
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Dimitri Budinger
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Joanne Ng
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Genetic Therapy Accelerator Centre, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - John Counsell
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Arlo Simpson
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Toni S Pearson
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032-3784, USA
- Department of Pediatrics, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43210, USA
- Department of Neurology, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43210, USA
| | - Simon Edvardson
- Department of Genetics, Hadassah, Hebrew University Medical Center, 9574869 Jerusalem, Israel
| | - Orly Elpeleg
- Department of Genetics, Hadassah, Hebrew University Medical Center, 9574869 Jerusalem, Israel
| | - Frances M Brodsky
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
| | - Gabriele Lignani
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Neurology, Great Ormond Street Hospital, London, WC1N 3JH, UK
| |
Collapse
|
3
|
Okafor O, Kim K. Cytotoxicity of Quantum Dots in Receptor-Mediated Endocytic and Pinocytic Pathways in Yeast. Int J Mol Sci 2024; 25:4714. [PMID: 38731933 PMCID: PMC11083673 DOI: 10.3390/ijms25094714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the promising applications of the use of quantum dots (QDs) in the biomedical field, the long-lasting effects of QDs on the cell remain poorly understood. To comprehend the mechanisms underlying the toxic effects of QDs in yeast, we characterized defects associated with receptor-mediated endocytosis (RME) as well as pinocytosis using Saccharomyces cerevisiae as a model in the presence of cadmium selenide/zinc sulfide (CdSe/ZnS) QDs. Our findings revealed that QDs led to an inefficient RME at the early, intermediate, and late stages of endocytic patch maturation at the endocytic site, with the prolonged lifespan of GFP fused yeast fimbrin (Sac6-GFP), a late marker of endocytosis. The transit of FM1-43, a lipophilic dye from the plasma membrane to the vacuole, was severely retarded in the presence of QDs. Finally, QDs caused an accumulation of monomeric red fluorescent protein fused carbamoyl phosphate synthetase 1 (mRFP-Cps1), a vacuolar lumen marker in the vacuole. In summary, the present study provides novel insights into the possible impact of CdSe/ZnS QDs on the endocytic machinery, enabling a deeper comprehension of QD toxicity.
Collapse
Affiliation(s)
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National, Springfield, MO 65897, USA;
| |
Collapse
|
4
|
Higashio H, Yokoyama T, Saino T. A convenient fluorimetry-based degranulation assay using RBL-2H3 cells. Biosci Biotechnol Biochem 2024; 88:181-188. [PMID: 37968134 DOI: 10.1093/bbb/zbad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
Type I hypersensitivity is triggered by mast cell degranulation, a stimulus-induced exocytosis of preformed secretory granules (SGs) containing various inflammatory mediators. The degree of degranulation is generally expressed as a percentage of secretory granule markers (such as β-hexosaminidase and histamine) released into the external solution, and considerable time and labor are required for the quantification of markers in both the supernatants and cell lysates. In this study, we developed a simple fluorimetry-based degranulation assay using rat basophilic leukemia (RBL-2H3) mast cells. During degranulation, the styryl dye FM1-43 in the external solution fluorescently labeled the newly exocytosed SGs, whose increase in intensity was successively measured using a fluorescence microplate reader. In addition to the rate of β-hexosaminidase secretion, the cellular FM1-43 intensity successfully represented the degree and kinetics of degranulation under various conditions, suggesting that this method facilitates multi-sample and/or multi-time-point analyses required for screening substances regulating mast cell degranulation.
Collapse
Affiliation(s)
- Hironori Higashio
- Department of Chemistry, Center for Liberal Arts and Sciences, Iwate Medical University, Yahaba, Iwate, Japan
| | - Takuya Yokoyama
- Division of Cell Biology, Department of Anatomy, Iwate Medical University, Yahaba, Iwate, Japan
| | - Tomoyuki Saino
- Division of Cell Biology, Department of Anatomy, Iwate Medical University, Yahaba, Iwate, Japan
| |
Collapse
|
5
|
Dubicka Z, Bojanowski MJ, Bijma J, Bickmeyer U. Mg-rich amorphous to Mg-low crystalline CaCO 3 pathway in foraminifera. Heliyon 2023; 9:e18331. [PMID: 37519760 PMCID: PMC10375801 DOI: 10.1016/j.heliyon.2023.e18331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/13/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Calcium carbonate minerals produced by marine organisms play a central role in the global carbon cycle and carbonate sedimentation, which influence the climate by regulating atmospheric CO2 levels. Foraminifera are important marine single-celled organisms that have produced calcite shells for over 300 million years. Here, we present new observations promoting our understanding for foraminiferal biocalcification by studying Amphistegina lessonii. We integrated in vivo confocal autofluorescence and dye fluorescence imaging with elemental analysis of the cell supporting the concept that the calcite shells of foraminifera are produced via deposition of intracellularly formed Mg-rich amorphous calcium carbonate (Mg-ACC) particles that transform into a stable mineral phase. This process is likely accompanied by the activity of endosymbiotic microalgae and seawater-derived endocytic vesicles that provide calcification substrates such as DIC, Ca2+, and Mg2+. The final transformation of semi-liquid amorphous nanoparticles into a crystalline shell was associated with Mg2+ liberation.
Collapse
Affiliation(s)
- Zofia Dubicka
- Ecological Chemistry, Alfred-Wegener-Institut Helmholtz-Zentrum für Polar- und Meeresforschung, Bremerhaven, 27-570, Germany
- University of Warsaw, Warsaw, 02-089, Poland
| | | | - Jelle Bijma
- Marine Biogeosciences, Alfred-Wegener-Institut Helmholtz-Zentrum für Polar- und Meeresforschung, Bremerhaven, 27-570, Germany
| | - Ulf Bickmeyer
- Ecological Chemistry, Alfred-Wegener-Institut Helmholtz-Zentrum für Polar- und Meeresforschung, Bremerhaven, 27-570, Germany
| |
Collapse
|
6
|
Potcoava M, Contini D, Zurawski Z, Huynh S, Mann C, Art J, Alford S. Live Cell Light Sheet Imaging with Low- and High-Spatial-Coherence Detection Approaches Reveals Spatiotemporal Aspects of Neuronal Signaling. J Imaging 2023; 9:121. [PMID: 37367469 PMCID: PMC10299414 DOI: 10.3390/jimaging9060121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Light sheet microscopy in live cells requires minimal excitation intensity and resolves three-dimensional (3D) information rapidly. Lattice light sheet microscopy (LLSM) works similarly but uses a lattice configuration of Bessel beams to generate a flatter, diffraction-limited z-axis sheet suitable for investigating subcellular compartments, with better tissue penetration. We developed a LLSM method for investigating cellular properties of tissue in situ. Neural structures provide an important target. Neurons are complex 3D structures, and signaling between cells and subcellular structures requires high resolution imaging. We developed an LLSM configuration based on the Janelia Research Campus design or in situ recording that allows simultaneous electrophysiological recording. We give examples of using LLSM to assess synaptic function in situ. In presynapses, evoked Ca2+ entry causes vesicle fusion and neurotransmitter release. We demonstrate the use of LLSM to measure stimulus-evoked localized presynaptic Ca2+ entry and track synaptic vesicle recycling. We also demonstrate the resolution of postsynaptic Ca2+ signaling in single synapses. A challenge in 3D imaging is the need to move the emission objective to maintain focus. We have developed an incoherent holographic lattice light-sheet (IHLLS) technique to replace the LLS tube lens with a dual diffractive lens to obtain 3D images of spatially incoherent light diffracted from an object as incoherent holograms. The 3D structure is reproduced within the scanned volume without moving the emission objective. This eliminates mechanical artifacts and improves temporal resolution. We focus on LLS and IHLLS applications and data obtained in neuroscience and emphasize increases in temporal and spatial resolution using these approaches.
Collapse
Affiliation(s)
- Mariana Potcoava
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Donatella Contini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Zachary Zurawski
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Spencer Huynh
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Christopher Mann
- Department of Applied Physics and Materials Science, Northern Arizona University, Flagstaff, AZ 86011, USA
- Center for Materials Interfaces in Research and Development, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Jonathan Art
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 South Wood Street, Rm 578 MC 512, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Chang HF, Schirra C, Pattu V, Krause E, Becherer U. Lytic granule exocytosis at immune synapses: lessons from neuronal synapses. Front Immunol 2023; 14:1177670. [PMID: 37275872 PMCID: PMC10233144 DOI: 10.3389/fimmu.2023.1177670] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Regulated exocytosis is a central mechanism of cellular communication. It is not only the basis for neurotransmission and hormone release, but also plays an important role in the immune system for the release of cytokines and cytotoxic molecules. In cytotoxic T lymphocytes (CTLs), the formation of the immunological synapse is required for the delivery of the cytotoxic substances such as granzymes and perforin, which are stored in lytic granules and released via exocytosis. The molecular mechanisms of their fusion with the plasma membrane are only partially understood. In this review, we discuss the molecular players involved in the regulated exocytosis of CTL, highlighting the parallels and differences to neuronal synaptic transmission. Additionally, we examine the strengths and weaknesses of both systems to study exocytosis.
Collapse
|
8
|
Mantilla CB, Ermilov LG, Greising SM, Gransee HM, Zhan WZ, Sieck GC. Electrophysiological effects of BDNF and TrkB signaling at type-identified diaphragm neuromuscular junctions. J Neurophysiol 2023; 129:781-792. [PMID: 36883761 PMCID: PMC10069962 DOI: 10.1152/jn.00015.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Previous studies show that synaptic quantal release decreases during repetitive stimulation, i.e., synaptic depression. Neurotrophin brain-derived neurotrophic factor (BDNF) enhances neuromuscular transmission via activation of tropomyosin-related kinase receptor B (TrkB). We hypothesized that BDNF mitigates synaptic depression at the neuromuscular junction and that the effect is more pronounced at type IIx and/or IIb fibers compared to type I or IIa fibers given the more rapid reduction in docked synaptic vesicles with repetitive stimulation. Rat phrenic nerve-diaphragm muscle preparations were used to determine the effect of BDNF on synaptic quantal release during repetitive stimulation at 50 Hz. An ∼40% decline in quantal release was observed during each 330-ms duration train of nerve stimulation (intratrain synaptic depression), and this intratrain decline was observed across repetitive trains (20 trains at 1/s repeated every 5 min for 30 min for 6 sets). BDNF treatment significantly enhanced quantal release at all fiber types (P < 0.001). BDNF treatment did not change release probability within a stimulation set but enhanced synaptic vesicle replenishment between sets. In agreement, synaptic vesicle cycling (measured using FM4-64 fluorescence uptake) was increased following BDNF [or neurotrophin-4 (NT-4)] treatment (∼40%; P < 0.05). Conversely, inhibiting BDNF/TrkB signaling with the tyrosine kinase inhibitor K252a and TrkB-IgG (which quenches endogenous BDNF or NT-4) decreased FM4-64 uptake (∼34% across fiber types; P < 0.05). The effects of BDNF were generally similar across all fiber types. We conclude that BDNF/TrkB signaling acutely enhances presynaptic quantal release and thereby may serve to mitigate synaptic depression and maintain neuromuscular transmission during repetitive activation.NEW & NOTEWORTHY Neurotrophin brain-derived neurotrophic factor (BDNF) enhances neuromuscular transmission via activation of tropomyosin-related kinase receptor B (TrkB). Rat phrenic nerve-diaphragm muscle preparations were used to determine the rapid effect of BDNF on synaptic quantal release during repetitive stimulation. BDNF treatment significantly enhanced quantal release at all fiber types. BDNF increased synaptic vesicle cycling (measured using FM4-64 fluorescence uptake); conversely, inhibiting BDNF/TrkB signaling decreased FM4-64 uptake.
Collapse
Affiliation(s)
- Carlos B Mantilla
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Leonid G Ermilov
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, United States
| | - Heather M Gransee
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Wen-Zhi Zhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Gary C Sieck
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
9
|
Bigge BM, Rosenthal NE, Avasthi P. Initial ciliary assembly in Chlamydomonas requires Arp2/3 complex-dependent endocytosis. Mol Biol Cell 2023; 34:ar24. [PMID: 36753382 PMCID: PMC10092647 DOI: 10.1091/mbc.e22-09-0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Ciliary assembly, trafficking, and regulation are dependent on microtubules, but the mechanisms of ciliary assembly also require the actin cytoskeleton. Here, we dissect subcellular roles of actin in ciliogenesis by focusing on actin networks nucleated by the Arp2/3 complex in the powerful ciliary model, Chlamydomonas. We find that the Arp2/3 complex is required for the initial stages of ciliary assembly when protein and membrane are in high demand but cannot yet be supplied from the Golgi complex. We provide evidence for Arp2/3 complex-dependent endocytosis of ciliary proteins, an increase in endocytic activity upon induction of ciliary growth, and relocalization of plasma membrane proteins to newly formed cilia.
Collapse
Affiliation(s)
- Brae M Bigge
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755; Anatomy and Cell Biology Department, University of Kansas Medical Center, Kansas City, KS 66103
| | - Nicholas E Rosenthal
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755; Anatomy and Cell Biology Department, University of Kansas Medical Center, Kansas City, KS 66103
| | - Prachee Avasthi
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755; Anatomy and Cell Biology Department, University of Kansas Medical Center, Kansas City, KS 66103
| |
Collapse
|
10
|
Landajuela A, Braun M, Martínez-Calvo A, Rodrigues CDA, Gomis Perez C, Doan T, Rudner DZ, Wingreen NS, Karatekin E. Membrane fission during bacterial spore development requires cellular inflation driven by DNA translocation. Curr Biol 2022; 32:4186-4200.e8. [PMID: 36041438 PMCID: PMC9730832 DOI: 10.1016/j.cub.2022.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/26/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022]
Abstract
Bacteria require membrane fission for both cell division and endospore formation. In Bacillus subtilis, sporulation initiates with an asymmetric division that generates a large mother cell and a smaller forespore that contains only a quarter of its genome. As the mother cell membranes engulf the forespore, a DNA translocase pumps the rest of the chromosome into the small forespore compartment, inflating it due to increased turgor. When the engulfing membrane undergoes fission, the forespore is released into the mother cell cytoplasm. The B. subtilis protein FisB catalyzes membrane fission during sporulation, but the molecular basis is unclear. Here, we show that forespore inflation and FisB accumulation are both required for an efficient membrane fission. Forespore inflation leads to higher membrane tension in the engulfment membrane than in the mother cell membrane, causing the membrane to flow through the neck connecting the two membrane compartments. Thus, the mother cell supplies some of the membrane required for the growth of the membranes surrounding the forespore. The oligomerization of FisB at the membrane neck slows the equilibration of membrane tension by impeding the membrane flow. This leads to a further increase in the tension of the engulfment membrane, promoting its fission through lysis. Collectively, our data indicate that DNA translocation has a previously unappreciated second function in energizing the FisB-mediated membrane fission under energy-limited conditions.
Collapse
Affiliation(s)
- Ane Landajuela
- Cellular and Molecular Physiology, Yale University, New Haven, CT, USA; Nanobiology Institute, Yale University, West Haven, CT, USA.
| | - Martha Braun
- Nanobiology Institute, Yale University, West Haven, CT, USA; Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA.
| | - Alejandro Martínez-Calvo
- Princeton Center for Theoretical Science, Princeton University, Princeton, NJ 08544, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | | | - Carolina Gomis Perez
- Cellular and Molecular Physiology, Yale University, New Haven, CT, USA; Nanobiology Institute, Yale University, West Haven, CT, USA
| | - Thierry Doan
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, Aix-Marseille Université-CNRS UMR7255, Marseilles, France
| | - David Z Rudner
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Ned S Wingreen
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Erdem Karatekin
- Cellular and Molecular Physiology, Yale University, New Haven, CT, USA; Nanobiology Institute, Yale University, West Haven, CT, USA; Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA; Université de Paris, Saints-Pères Paris Institute for the Neurosciences (SPPIN), Centre National de la Recherche Scientifique (CNRS), 75006 Paris, France.
| |
Collapse
|
11
|
Wang X, Sun M, Chan CY, Wu LG. Phospholipase A 2-based probes to study vesicle trafficking. CELL REPORTS METHODS 2022; 2:100206. [PMID: 35497501 PMCID: PMC9046444 DOI: 10.1016/j.crmeth.2022.100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Vesicle exo- and endocytosis mediate important biological functions, including synaptic transmission. In this issue of Cell Reports Methods, Seong J. An et al. found that the fluorescently tagged C2 domain of phospholipase A2 binds to membrane phosphatidylcholine and thus labels vesicle membrane, allowing for super-resolution and electron microscopic visualization of vesicle trafficking.
Collapse
Affiliation(s)
- Xin Wang
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bldg. 35, Rm. 2B-1012, Bethesda, MD 20892, USA
| | - Min Sun
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bldg. 35, Rm. 2B-1012, Bethesda, MD 20892, USA
| | - Chung Yu Chan
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bldg. 35, Rm. 2B-1012, Bethesda, MD 20892, USA
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bldg. 35, Rm. 2B-1012, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Rodrigues PV, de Godoy JVP, Bosque BP, Amorim Neto DP, Tostes K, Palameta S, Garcia-Rosa S, Tonoli CCC, de Carvalho HF, de Castro Fonseca M. Transcellular propagation of fibrillar α-synuclein from enteroendocrine to neuronal cells requires cell-to-cell contact and is Rab35-dependent. Sci Rep 2022; 12:4168. [PMID: 35264710 PMCID: PMC8907230 DOI: 10.1038/s41598-022-08076-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/02/2022] [Indexed: 01/15/2023] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative condition featured by motor dysfunction, death of midbrain dopaminergic neurons and accumulation of α-synuclein (αSyn) aggregates. Growing evidence suggests that PD diagnosis happens late in the disease progression and that the pathology may originate much earlier in the enteric nervous system (ENS) before advancing to the brain, via autonomic fibers. It was recently described that a specific cell type from the gut epithelium named enteroendocrine cells (EECs) possess many neuron-like properties including αSyn expression. By facing the gut lumen and being directly connected with αSyn-containing enteric neurons in a synaptic manner, EECs form a neural circuit between the gastrointestinal tract and the ENS, thereby being a possible key player in the outcome of PD in the gut. We have characterized the progression and the cellular mechanisms involved in αSyn pre-formed fibrils (PFFs) transfer from EECs to neuronal cells. We show that brain organoids efficiently internalize αSyn PFF seeds which triggers the formation of larger intracellular inclusions. In addition, in the enteroendocrine cell line STC-1 and in the neuronal cell line SH-SY5Y, αSyn PFFs induced intracellular calcium (Ca2+) oscillations on an extracellular Ca2+ source-dependent manner and triggered αSyn fibrils internalization by endocytosis. We characterized the spread of αSyn PFFs from enteroendocrine to neuronal cells and showed that this process is dependent on physical cell-to-cell contact and on Rab35 GTPase. Lastly, inhibition of Rab35 increases the clearance of αSyn fibrils by redirecting them to the lysosomal compartment. Therefore, our results reveal mechanisms that contribute to the understanding of how seeded αSyn fibrils promote the progression of αSyn pathology from EECs to neuronal cells shifting the focus of PD etiology to the ENS.
Collapse
Affiliation(s)
- Paulla Vieira Rodrigues
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - João Vitor Pereira de Godoy
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Beatriz Pelegrini Bosque
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Dionísio Pedro Amorim Neto
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil.,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Katiane Tostes
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | - Soledad Palameta
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | - Sheila Garcia-Rosa
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | - Celisa Caldana Costa Tonoli
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil
| | | | - Matheus de Castro Fonseca
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio), 10000 Giuseppe Maximo Scolfaro St., Campinas, São Paulo, 13083-100, Brazil. .,Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil. .,Laboratory of Sarkis Mazmanian, Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA, USA.
| |
Collapse
|
13
|
Revisiting Thin-Layer Electrochemistry in a Chip-Type Cell for the Study of Electro-organic Reactions. Anal Chem 2021; 94:1248-1255. [PMID: 34964606 DOI: 10.1021/acs.analchem.1c04467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It is important but challenging to elucidate the electrochemical reaction mechanisms of organic compounds using electroanalytical methods. Particularly, a rapid and straightforward method that provides information on reaction intermediates or other key electrochemical parameters may be useful. In this work, we exploited the advantages of classic thin-layer electrochemistry to develop a thin-layer electroanalysis microchip (TEAM). The TEAM provided better-resolved voltammetric peaks than under semi-infinite diffusion conditions owing to its small height. Importantly, rapid and accurate determination of the number of electrons transferred, n, was enabled by mechanically confining the microliter-scale volume analyte at the electrode, while securing ionic conduction using polyelectrolyte gels. The performance of the TEAM was validated using voltammetry and coulometry of standard redox couples. Utilizing the TEAM, a (spectro)electrochemical analysis of FM 1-43, an organic dye widely used in neuroscience, was successfully performed. Moreover, the TEAM was applied to study the electrochemical oxidation mechanism of pivanilides and alkyltrifluoroborate salts with different substituents and solvents. This work suggests that TEAM is a promising tool to provide invaluable mechanistic information and promote the rational design of electrosynthetic strategies.
Collapse
|
14
|
Rituper B, Guček A, Lisjak M, Gorska U, Šakanović A, Bobnar ST, Lasič E, Božić M, Abbineni PS, Jorgačevski J, Kreft M, Verkhratsky A, Platt FM, Anderluh G, Stenovec M, Božič B, Coorssen JR, Zorec R. Vesicle cholesterol controls exocytotic fusion pore. Cell Calcium 2021; 101:102503. [PMID: 34844123 DOI: 10.1016/j.ceca.2021.102503] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022]
Abstract
In some lysosomal storage diseases (LSD) cholesterol accumulates in vesicles. Whether increased vesicle cholesterol affects vesicle fusion with the plasmalemma, where the fusion pore, a channel between the vesicle lumen and the extracellular space, is formed, is unknown. Super-resolution microscopy revealed that after stimulation of exocytosis, pituitary lactotroph vesicles discharge cholesterol which transfers to the plasmalemma. Cholesterol depletion in lactotrophs and astrocytes, both exhibiting Ca2+-dependent exocytosis regulated by distinct Ca2+sources, evokes vesicle secretion. Although this treatment enhanced cytosolic levels of Ca2+ in lactotrophs but decreased it in astrocytes, this indicates that cholesterol may well directly define the fusion pore. In an attempt to explain this mechanism, a new model of cholesterol-dependent fusion pore regulation is proposed. High-resolution membrane capacitance measurements, used to monitor fusion pore conductance, a parameter related to fusion pore diameter, confirm that at resting conditions reducing cholesterol increases, while enrichment with cholesterol decreases the conductance of the fusion pore. In resting fibroblasts, lacking the Npc1 protein, a cellular model of LSD in which cholesterol accumulates in vesicles, the fusion pore conductance is smaller than in controls, showing that vesicle cholesterol controls fusion pore and is relevant for pathophysiology of LSD.
Collapse
Affiliation(s)
- Boštjan Rituper
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Alenka Guček
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Marjeta Lisjak
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Urszula Gorska
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Aleksandra Šakanović
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Saša Trkov Bobnar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Eva Lasič
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Mićo Božić
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Prabhodh S Abbineni
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, United States of America
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Alexei Verkhratsky
- Celica Biomedical, 1000, Ljubljana, Slovenia; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, United Kingdom
| | - Gregor Anderluh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Bojan Božič
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Jens R Coorssen
- Department of Health Sciences, Faculty of Applied Health Sciences and Department of Biological Sciences, Faculty of Mathematics & Science, Brock University, St Catherine's, Ontario, Canada
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia.
| |
Collapse
|
15
|
Wakatsuki S, Takahashi Y, Shibata M, Adachi N, Numakawa T, Kunugi H, Araki T. Small noncoding vault RNA modulates synapse formation by amplifying MAPK signaling. J Cell Biol 2021; 220:211679. [PMID: 33439240 PMCID: PMC7809882 DOI: 10.1083/jcb.201911078] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 09/04/2020] [Accepted: 12/02/2020] [Indexed: 12/30/2022] Open
Abstract
The small noncoding vault RNA (vtRNA) is a component of the vault complex, a ribonucleoprotein complex found in most eukaryotes. Emerging evidence suggests that vtRNAs may be involved in the regulation of a variety of cellular functions when unassociated with the vault complex. Here, we demonstrate a novel role for vtRNA in synaptogenesis. Using an in vitro synapse formation model, we show that murine vtRNA (mvtRNA) promotes synapse formation by modulating the MAPK signaling pathway. mvtRNA is transported to the distal region of neurites as part of the vault complex. Interestingly, mvtRNA is released from the vault complex in the neurite by a mitotic kinase Aurora-A–dependent phosphorylation of MVP, a major protein component of the vault complex. mvtRNA binds to and activates MEK1 and thereby enhances MEK1-mediated ERK activation in neurites. These results suggest the existence of a regulatory mechanism of the MAPK signaling pathway by vtRNAs as a new molecular basis for synapse formation.
Collapse
Affiliation(s)
- Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoko Takahashi
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Megumi Shibata
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Naoki Adachi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo, Japan
| | - Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
16
|
Lemière J, Ren Y, Berro J. Rapid adaptation of endocytosis, exocytosis and eisosomes after an acute increase in membrane tension in yeast cells. eLife 2021; 10:62084. [PMID: 33983119 PMCID: PMC9045820 DOI: 10.7554/elife.62084] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 05/12/2021] [Indexed: 11/13/2022] Open
Abstract
During clathrin-mediated endocytosis (CME) in eukaryotes, actin assembly is required to overcome large membrane tension and turgor pressure. However, the molecular mechanisms by which the actin machinery adapts to varying membrane tension remain unknown. In addition, how cells reduce their membrane tension when they are challenged by hypotonic shocks remains unclear. We used quantitative microscopy to demonstrate that cells rapidly reduce their membrane tension using three parallel mechanisms. In addition to using their cell wall for mechanical protection, yeast cells disassemble eisosomes to buffer moderate changes in membrane tension on a minute time scale. Meanwhile, a temporary reduction in the rate of endocytosis for 2–6 min and an increase in the rate of exocytosis for at least 5 min allow cells to add large pools of membrane to the plasma membrane. We built on these results to submit the cells to abrupt increases in membrane tension and determine that the endocytic actin machinery of fission yeast cells rapidly adapts to perform CME. Our study sheds light on the tight connection between membrane tension regulation, endocytosis, and exocytosis.
Collapse
Affiliation(s)
- Joël Lemière
- Department of Molecular Biophysics and Biochemistry, Department of Cell Biology, Yale University, New Haven, United States
| | - Yuan Ren
- Department of Molecular Biophysics and Biochemistry, Department of Cell Biology, Yale University, New Haven, United States
| | - Julien Berro
- Department of Molecular Biophysics and Biochemistry, Department of Cell Biology, Yale University, New Haven, United States
| |
Collapse
|
17
|
A surfactant polymer wound dressing protects human keratinocytes from inducible necroptosis. Sci Rep 2021; 11:4357. [PMID: 33623080 PMCID: PMC7902632 DOI: 10.1038/s41598-021-82260-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic wounds show necroptosis from which keratinocytes must be protected to enable appropriate wound re-epithelialization and closure. Poloxamers, a class of synthetic triblock copolymers, are known to be effective against plasma membrane damage (PMD). The purpose of this study is to evaluate the efficacy of a specific poloxamer, surfactant polymer dressing (SPD), which is currently used clinically as wound care dressing, against PMD in keratinocytes. Triton X-100 (TX100) at sub-lytic concentrations caused PMD as demonstrated by the efflux of calcein and by the influx of propidium iodide and FM1-43. TX100, an inducer of necroptosis, led to mitochondrial fragmentation, depletion of nuclear HMGB1, and activation of signaling complex associated with necroptosis (i.e., activation of RIP3 and phosphorylation of MLKL). All responses following exposure of human keratinocytes to TX100 were attenuated by pre- or co-treatment with SPD (100 mg/ml). The activation and translocation of phospho-MLKL to the plasma membrane, taken together with depletion of nuclear HMGB1, characterized the observed cell death as necroptosis. Thus, our findings show that TX100-induced plasma membrane damage and death by necroptosis were both attenuated by SPD, allowing keratinocyte survival. The significance of such protective effects of SPD on keratinocytes in wound re-epithelialization and closure warrant further studies.
Collapse
|
18
|
Zhang H, Aoki T, Hatano K, Kabayama K, Nakagawa M, Fukase K, Okamura Y. Porous nanosheet wrapping for live imaging of suspension cells. J Mater Chem B 2020; 6:6622-6628. [PMID: 31999284 DOI: 10.1039/c8tb01943f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the field of cell imaging, it is still a practical challenge to obtain the high quality live imaging of suspension cells, mainly due to undesirable cell movement in the imaging field during observation. This study describes a porous nanosheet wrapping method to noninvasively immobilize suspension cells for their live imaging. Perforated nanopores are fabricated on a nanosheet to enable the addition of external chemicals to cells, ranging from small molecules to macromolecules. Through several case studies, such as the live imaging of membrane staining of liposomes, transferrin endocytosis of B cells, and activation of platelets, it is verified that the confined space made by the nanosheet could provide a hydrodynamically stable environment for suspension cells, even if an aqueous stimulus is added through the nanopores in a static or a flowing condition. With this method, the live imaging of the whole activation process on a specific suspension cell in the imaging field is achieved, which is not feasible with the existing cell immobilization methods. This study suggests that the method of porous nanosheet wrapping will facilitate the visualization of the dynamic functions of suspension cells.
Collapse
Affiliation(s)
- Hong Zhang
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan.
| | - Takuto Aoki
- Course of Applied Science, Graduate School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
| | - Kanae Hatano
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Masaru Nakagawa
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Yosuke Okamura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan. and Course of Applied Science, Graduate School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
| |
Collapse
|
19
|
Avila R, Tamariz E, Medina-Villalobos N, Andilla J, Marsal M, Loza-Alvarez P. Effects of near infrared focused laser on the fluorescence of labelled cell membrane. Sci Rep 2018; 8:17674. [PMID: 30518772 PMCID: PMC6281678 DOI: 10.1038/s41598-018-36010-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/09/2018] [Indexed: 11/12/2022] Open
Abstract
Near infrared (NIR) laser light can have important reactions on live cells. For example, in a macroscopic scale, it is used therapeutically to reduce inflammation and in a single-cell scale, NIR lasers have been experimentally used to guide neuronal growth. However, little is known about how NIR lasers produce such behaviours on cells. In this paper we report effects of focussing a continuous wave 810-nm wavelength laser on in vivo 3T3 cells plasma membrane. Cell membranes were labelled with FM 4-64, a dye that fluoresces when associated to membrane lipids. Confocal microscopy was used to image cell membranes and perform fluorescence recovery after photobleaching (FRAP) experiments. We found that the NIR laser produces an increase of the fluorescence intensity at the location of laser spot. This intensity boost vanishes once the laser is turned off. The mean fluorescence increase, calculated over 75 independent measurements, equals 19%. The experiments reveal that the fluorescence rise is a growing function of the laser power. This dependence is well fitted with a square root function. The FRAP, when the NIR laser is acting on the cell, is twice as large as when the NIR laser is off, and the recovery time is 5 times longer. Based on the experimental evidence and a linear fluorescence model, it is shown that the NIR laser provokes a rise in the number of molecular associations dye-lipid. The results reported here may be a consequence of a combination of induced increments in membrane fluidity and exocytosis.
Collapse
Affiliation(s)
- Remy Avila
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México (UNAM), A. P. 1-1010, Juriquilla, 76000, Querétaro, Mexico. .,ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain.
| | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenicda Luis Castelazo Ayala s/n, Xalapa, 91190, Veracruz, Mexico
| | - Norma Medina-Villalobos
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain.,Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenicda Luis Castelazo Ayala s/n, Xalapa, 91190, Veracruz, Mexico
| | - Jordi Andilla
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain
| | - María Marsal
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain
| | - Pablo Loza-Alvarez
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain
| |
Collapse
|
20
|
Nakanishi H, Kurima K, Pan B, Wangemann P, Fitzgerald TS, Géléoc GS, Holt JR, Griffith AJ. Tmc2 expression partially restores auditory function in a mouse model of DFNB7/B11 deafness caused by loss of Tmc1 function. Sci Rep 2018; 8:12125. [PMID: 30108230 PMCID: PMC6092339 DOI: 10.1038/s41598-018-29709-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/03/2018] [Indexed: 11/19/2022] Open
Abstract
Mouse Tmc1 and Tmc2 are required for sensory transduction in cochlear and vestibular hair cells. Homozygous Tmc1∆/∆ mice are deaf, Tmc2∆/∆ mice have normal hearing, and double homozygous Tmc1∆/∆; Tmc2∆/∆ mice have deafness and profound vestibular dysfunction. These phenotypes are consistent with their different spatiotemporal expression patterns. Tmc1 expression is persistent in cochlear and vestibular hair cells, whereas Tmc2 expression is transient in cochlear hair cells but persistent in vestibular hair cells. On the basis of these findings, we hypothesized that persistent Tmc2 expression in mature cochlear hair cells could restore auditory function in Tmc1∆/∆ mice. To express Tmc2 in mature cochlear hair cells, we generated a transgenic mouse line, Tg[PTmc1::Tmc2], in which Tmc2 cDNA is expressed under the control of the Tmc1 promoter. The Tg[PTmc1::Tmc2] transgene slightly but significantly restored hearing in young Tmc1∆/∆ mice, though hearing thresholds were elevated with age. The elevation of hearing thresholds was associated with deterioration of sensory transduction in inner hair cells and loss of outer hair cell function. Although sensory transduction was retained in outer hair cells, their stereocilia eventually degenerated. These results indicate distinct roles and requirements for Tmc1 and Tmc2 in mature cochlear hair cells.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Hair Cells, Auditory/cytology
- Hair Cells, Auditory/metabolism
- Hair Cells, Auditory/pathology
- Hair Cells, Auditory/ultrastructure
- Hair Cells, Vestibular/metabolism
- Hearing Loss, Sensorineural/diagnosis
- Hearing Loss, Sensorineural/genetics
- Hearing Loss, Sensorineural/pathology
- Hearing Tests
- Homozygote
- Humans
- Mechanotransduction, Cellular
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Knockout
- Microscopy, Electron, Scanning
- Mutation
- Patch-Clamp Techniques
- Promoter Regions, Genetic/genetics
- Stereocilia/pathology
- Stereocilia/ultrastructure
Collapse
Affiliation(s)
- Hiroshi Nakanishi
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, 20892, USA
| | - Kiyoto Kurima
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, 20892, USA
| | - Bifeng Pan
- Departments of Otolaryngology and Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Philine Wangemann
- Anatomy and Physiology Department, Kansas State University, Manhattan, Kansas, 66506, USA
| | - Tracy S Fitzgerald
- Mouse Auditory Testing Core Facility, NIDCD, NIH, Bethesda, Maryland, 20892, USA
| | - Gwenaëlle S Géléoc
- Departments of Otolaryngology and Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Andrew J Griffith
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
21
|
Complexin Mutants Reveal Partial Segregation between Recycling Pathways That Drive Evoked and Spontaneous Neurotransmission. J Neurosci 2017; 37:383-396. [PMID: 28077717 DOI: 10.1523/jneurosci.1854-16.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/21/2022] Open
Abstract
Synaptic vesicles fuse at morphological specializations in the presynaptic terminal termed active zones (AZs). Vesicle fusion can occur spontaneously or in response to an action potential. Following fusion, vesicles are retrieved and recycled within nerve terminals. It is still unclear whether vesicles that fuse spontaneously or following evoked release share similar recycling mechanisms. Genetic deletion of the SNARE-binding protein complexin dramatically increases spontaneous fusion, with the protein serving as the synaptic vesicle fusion clamp at Drosophila synapses. We examined synaptic vesicle recycling pathways at complexin null neuromuscular junctions, where spontaneous release is dramatically enhanced. We combined loading of the lipophilic dye FM1-43 with photoconversion, electron microscopy, and electrophysiology to monitor evoked and spontaneous recycling vesicle pools. We found that the total number of recycling vesicles was equal to those retrieved through spontaneous and evoked pools, suggesting that retrieval following fusion is partially segregated for spontaneous and evoked release. In addition, the kinetics of FM1-43 destaining and synaptic depression measured in the presence of the vesicle-refilling blocker bafilomycin indicated that spontaneous and evoked recycling pools partially intermix during the release process. Finally, FM1-43 photoconversion combined with electron microscopy analysis indicated that spontaneous recycling preferentially involves synaptic vesicles in the vicinity of AZs, whereas vesicles recycled following evoked release involve a larger intraterminal pool. Together, these results suggest that spontaneous and evoked vesicles use separable recycling pathways and then partially intermix during subsequent rounds of fusion. SIGNIFICANCE STATEMENT Neurotransmitter release involves fusion of synaptic vesicles with the plasma membrane in response to an action potential, or spontaneously in the absence of stimulation. Upon fusion, vesicles are retrieved and recycled, and it is unclear whether recycling pathways for evoked and spontaneous vesicles are segregated after fusion. We addressed this question by taking advantage of preparations lacking the synaptic protein complexin, which have elevated spontaneous release that enables reliable tracking of the spontaneous recycling pool. Our results suggest that spontaneous and evoked recycling pathways are segregated during the retrieval process but can partially intermix during stimulation.
Collapse
|
22
|
Pathak-Sharma S, Zhang X, Lam JGT, Weisleder N, Seveau SM. High-Throughput Microplate-Based Assay to Monitor Plasma Membrane Wounding and Repair. Front Cell Infect Microbiol 2017; 7:305. [PMID: 28770170 PMCID: PMC5509797 DOI: 10.3389/fcimb.2017.00305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
The plasma membrane of mammalian cells is susceptible to disruption by mechanical and biochemical damages that frequently occur within tissues. Therefore, efficient and rapid repair of the plasma membrane is essential for maintaining cellular homeostasis and survival. Excessive damage of the plasma membrane and defects in its repair are associated with pathological conditions such as infections, muscular dystrophy, heart failure, diabetes, and lung and neurodegenerative diseases. The molecular events that remodel the plasma membrane during its repair remain poorly understood. In the present work, we report the development of a quantitative high-throughput assay that monitors the efficiency of the plasma membrane repair in real time using a sensitive microplate reader. In this assay, the plasma membrane of living cells is perforated by the bacterial pore-forming toxin listeriolysin O and the integrity and recovery of the membrane are monitored at 37°C by measuring the fluorescence intensity of the membrane impermeant dye propidium iodide. We demonstrate that listeriolysin O causes dose-dependent plasma membrane wounding and activation of the cell repair machinery. This assay was successfully applied to cell types from different origins including epithelial and muscle cells. In conclusion, this high-throughput assay provides a novel opportunity for the discovery of membrane repair effectors and the development of new therapeutic compounds that could target membrane repair in various pathological processes, from degenerative to infectious diseases.
Collapse
Affiliation(s)
- Sarika Pathak-Sharma
- Department of Microbial Infection and Immunity, The Ohio State University Medical CenterColumbus, OH, United States
| | - Xiaoli Zhang
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University Medical CenterColumbus, OH, United States
| | - Jonathan G T Lam
- Department of Microbial Infection and Immunity, The Ohio State University Medical CenterColumbus, OH, United States.,Department of Microbiology, The Ohio State UniversityColumbus, OH, United States
| | - Noah Weisleder
- Department of Physiology and Cell Biology, The Ohio State University Medical Center, Davis Heart and Lung Research InstituteColumbus, OH, United States
| | - Stephanie M Seveau
- Department of Microbial Infection and Immunity, The Ohio State University Medical CenterColumbus, OH, United States.,Center for Microbial Infection Biology, The Ohio State University Medical CenterColumbus, OH, United States
| |
Collapse
|
23
|
Lazarenko RM, DelBove CE, Strothman CE, Zhang Q. Ammonium chloride alters neuronal excitability and synaptic vesicle release. Sci Rep 2017; 7:5061. [PMID: 28698583 PMCID: PMC5505971 DOI: 10.1038/s41598-017-05338-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/12/2017] [Indexed: 01/28/2023] Open
Abstract
Genetically encoded pH-sensors are widely used in studying cell membrane trafficking and membrane protein turnover because they render exo-/endocytosis-associated pH changes to fluorescent signals. For imaging and analysis purposes, high concentration ammonium chloride is routinely used to alkalize intracellular membrane compartments under the assumption that it does not cause long-term effects on cellular processes being studied like neurotransmission. However, pathological studies about hyperammonemia have shown that ammonium is toxic to brain cells especially astrocytes and neurons. Here, we focus on ammonium’s physiological impacts on neurons including membrane potential, cytosolic Ca2+ and synaptic vesicles. We have found that extracellularly applied ammonium chloride as low as 5 mM causes intracellular Ca2+-increase and a reduction of vesicle release even after washout. The often-used 50 mM ammonium chloride causes more extensive and persistent changes, including membrane depolarization, prolonged elevation of intracellular Ca2+ and diminution of releasable synaptic vesicles. Our findings not only help to bridge the discrepancies in previous studies about synaptic vesicle release using those pH-sensors or other vesicle specific reporters, but also suggest an intriguing relationship between intracellular pH and neurotransmission.
Collapse
Affiliation(s)
- Roman M Lazarenko
- Department of Pharmacology, Vanderbilt University, 23rd Avenue South at Pierce Street, Nashville, TN, 37232, USA
| | - Claire E DelBove
- Department of Pharmacology, Vanderbilt University, 23rd Avenue South at Pierce Street, Nashville, TN, 37232, USA
| | - Claire E Strothman
- Department of Pharmacology, Vanderbilt University, 23rd Avenue South at Pierce Street, Nashville, TN, 37232, USA
| | - Qi Zhang
- Department of Pharmacology, Vanderbilt University, 23rd Avenue South at Pierce Street, Nashville, TN, 37232, USA.
| |
Collapse
|
24
|
Torres VI, Inestrosa NC. Vertebrate Presynaptic Active Zone Assembly: a Role Accomplished by Diverse Molecular and Cellular Mechanisms. Mol Neurobiol 2017; 55:4513-4528. [PMID: 28685386 DOI: 10.1007/s12035-017-0661-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/14/2017] [Indexed: 01/22/2023]
Abstract
Among all the biological systems in vertebrates, the central nervous system (CNS) is the most complex, and its function depends on specialized contacts among neurons called synapses. The assembly and organization of synapses must be exquisitely regulated for a normal brain function and network activity. There has been a tremendous effort in recent decades to understand the molecular and cellular mechanisms participating in the formation of new synapses and their organization, maintenance, and regulation. At the vertebrate presynapses, proteins such as Piccolo, Bassoon, RIM, RIM-BPs, CAST/ELKS, liprin-α, and Munc13 are constant residents and participate in multiple and dynamic interactions with other regulatory proteins, which define network activity and normal brain function. Here, we review the function of these active zone (AZ) proteins and diverse factors involved in AZ assembly and maintenance, with an emphasis on axonal trafficking of precursor vesicles, protein homo- and hetero-oligomeric interactions as a mechanism of AZ trapping and stabilization, and the role of F-actin in presynaptic assembly and its modulation by Wnt signaling.
Collapse
Affiliation(s)
- Viviana I Torres
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
25
|
Fedorovich S, Hofmeijer J, van Putten MJAM, le Feber J. Reduced Synaptic Vesicle Recycling during Hypoxia in Cultured Cortical Neurons. Front Cell Neurosci 2017; 11:32. [PMID: 28261063 PMCID: PMC5311063 DOI: 10.3389/fncel.2017.00032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/01/2017] [Indexed: 01/08/2023] Open
Abstract
Improvement of neuronal recovery in the ischemic penumbra, an area around the core of a brain infarct with some remaining perfusion, has a large potential for the development of therapy against acute ischemic stroke. However, mechanisms that lead to either recovery or secondary damage in the penumbra largely remain unclear. Recent studies in cultured networks of cortical neurons showed that failure of synaptic transmission (referred to as synaptic failure) is a critical factor in the penumbral area, but the mechanisms that lead to synaptic failure are still under investigation. Here we used a Styryl dye, FM1-43, to quantify endocytosis and exocytosis in cultures of rat cortical neurons under normoxic and hypoxic conditions. Hypoxia in cultured cortical networks rapidly depressed endocytosis and, to a lesser extent, exocytosis. These findings support electrophysiological findings that synaptic failure occurs quickly after the induction of hypoxia, and confirms that the failing processes are at least in part presynaptic.
Collapse
Affiliation(s)
- Sergei Fedorovich
- Laboratory of Biophysics and Cellular Engineering, Institute of Biophysics and Cell Engineering, National Academy of Sciences of Belarus Minsk, Belarus
| | - Jeannette Hofmeijer
- Clinical Neurophysiology, University of TwenteEnschede, Netherlands; Department of Neurology, Rijnstate HospitalArnhem, Netherlands
| | - Michel J A M van Putten
- Clinical Neurophysiology, University of TwenteEnschede, Netherlands; Department of Neurology and Clinical Neurophysiology, Medisch Spectrum TwenteEnschede, Netherlands
| | - Joost le Feber
- Clinical Neurophysiology, University of Twente Enschede, Netherlands
| |
Collapse
|
26
|
Balseiro-Gomez S, Flores JA, Acosta J, Ramirez-Ponce MP, Ales E. Transient fusion ensures granule replenishment to enable repeated release after IgE-mediated mast cell degranulation. J Cell Sci 2016; 129:3989-4000. [PMID: 27624612 DOI: 10.1242/jcs.194340] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/07/2016] [Indexed: 11/20/2022] Open
Abstract
To ensure normal immune function, mast cells employ different pathways to release mediators. Here, we report a thus far unknown capacity of mast cells to recycle and reuse secretory granules after an antigen-evoked degranulation process under physiological conditions; this phenomenon involves the existence of a recycling secretory granule pool that is available for release in a short time scale. Rapid endocytic modes contributed to the recycling of ∼60% of the total secretory granule population, which involved kiss-and-run and cavicapture mechanisms, causing retention of the intragranular matrix. We found the presence of normal-size granules and giant actomyosin- and dynamin-dependent granules, which were characterized by large quantal content. These large structures allowed the recovered mast cells to release a large amount of 5-HT, compensating for the decrease in the number of exocytosed secretory granules. This work uncovers a new physiological role of the exo-endocytosis cycle in the immunological plasticity of mast cells and reveals a new property of their biological secretion.
Collapse
Affiliation(s)
- Santiago Balseiro-Gomez
- Departamento Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de 41009 Sevilla, Spain
| | - Juan A Flores
- Departamento Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de 41009 Sevilla, Spain
| | - Jorge Acosta
- Departamento Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de 41009 Sevilla, Spain
| | - M Pilar Ramirez-Ponce
- Departamento Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de 41009 Sevilla, Spain
| | - Eva Ales
- Departamento Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de 41009 Sevilla, Spain
| |
Collapse
|
27
|
Functional Nanoscale Imaging of Synaptic Vesicle Cycling with Superfast Fixation. Methods Mol Biol 2016. [PMID: 27515092 DOI: 10.1007/978-1-4939-6352-2_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Functional imaging is the measurement of structural changes during an ongoing physiological process over time. In many cases, functional imaging has been implemented by tracking a fluorescent signal in live imaging sessions. Electron microscopy, however, excludes live imaging which has hampered functional imaging approaches on the ultrastructural level. This barrier was broken with the introduction of superfast fixation. Superfast fixation is capable of stopping and fixing membrane traffic at sufficient speed to capture a physiological process at a distinct functional state. Applying superfast fixation at sequential time points allows tracking of membrane traffic in a step-by-step fashion.This technique has been applied to track labeled endocytic vesicles at central synapses as they pass through the synaptic vesicle cycle. At synapses, neurotransmitter is released from synaptic vesicles (SVs) via fast activity-dependent exocytosis. Exocytosis is coupled to fast endocytosis that retrieves SVs components from the plasma membrane shortly after release. Fluorescent FM dyes that bind to the outer leaflet of the plasma membrane enter the endocytic vesicle during membrane retrieval and remain trapped in endocytic vesicles have been widely used to study SV exo-endocytic cycling in live imaging sessions. FM dyes can also be photoconverted into an electron-dense diaminobenzidine polymer which allows the investigation of SV cycling in the electron microscope. The combination of FM labeling with superfast fixation made it possible to track the fine structure of endocytic vesicles at 1 s intervals. Because this combination is not specialized to SV cycling, many other cellular processes can be studied. Furthermore, the technique is easy to set up and cost effective.This chapter describes activity-dependent FM dye labeling of SVs in cultured hippocampal neurons, superfast microwave-assisted fixation, photoconversion of the fluorescent endocytic vesicles, and the analysis of individual synapses after serial section 3D reconstruction of individual synapses from electron micrographs.
Collapse
|
28
|
Liu J, Liu W, Yang J. ATP-containing vesicles in stria vascular marginal cell cytoplasms in neonatal rat cochlea are lysosomes. Sci Rep 2016; 6:20903. [PMID: 26864824 PMCID: PMC4750035 DOI: 10.1038/srep20903] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 01/13/2016] [Indexed: 01/03/2023] Open
Abstract
We confirmed that ATP is released from cochlear marginal cells in the stria vascular but the cell organelle in which ATP stores was not identified until now. Thus, we studied the ATP-containing cell organelles and suggest that these are lysosomes. Primary cultures of marginal cells of Sprague-Dawley rats aged 1-3 days was established. Vesicles within marginal cells stained with markers were identified under confocal laser scanning microscope and transmission electron microscope (TEM). Then ATP release from marginal cells was measured after glycyl-L-phenylalanine-ß- naphthylamide (GPN) treatment using a bioluminescent assay. Quinacrine-stained granules within marginal cells were labeled with LysoTracker, a lysosome tracer, and lysosomal-associated membrane protein 1(LAMP1), but not labeled with the mitochondrial tracer MitoTracker. Furthermore, LysoTracker-labelled puncta showed accumulation of Mant-ATP, an ATP analog. Treatment with 200 μM GPN quenched fluorescently labeled puncta after incubation with LysoTracker or quinacrine, but not MitoTracker. Quinacrine-labeled organelles observed by TEM were lysosomes, and an average 27.7 percent increase in ATP luminescence was observed in marginal cells extracellular fluid after GPN treatment. ATP-containing vesicles in cochlear marginal cells of the stria vascular from neonatal rats are likely lysosomes. ATP release from marginal cells may be via Ca(2+)-dependent lysosomal exocytosis.
Collapse
Affiliation(s)
- Jun Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Jiaotong University School of Medicine Ear Institute, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
- Department of Otorhinolaryngology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenjing Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Jiaotong University School of Medicine Ear Institute, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Jiaotong University School of Medicine Ear Institute, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| |
Collapse
|
29
|
López-Duarte I, Chairatana P, Wu Y, Pérez-Moreno J, Bennett PM, Reeve JE, Boczarow I, Kaluza W, Hosny NA, Stranks SD, Nicholas RJ, Clays K, Kuimova MK, Anderson HL. Thiophene-based dyes for probing membranes. Org Biomol Chem 2015; 13:3792-802. [PMID: 25703541 DOI: 10.1039/c4ob02507e] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We report the synthesis of four new cationic dipolar push–pull dyes, together with an evaluation of their photophysical and photobiological characteristics pertinent to imaging membranes by fluorescence and second harmonic generation (SHG). All four dyes consist of an N,N-diethylaniline electron-donor conjugated to a pyridinium electron-acceptor via a thiophene bridge, with either vinylene (–CH=CH–) or ethynylene (–C≡C–) linking groups, and with either singly-charged or doubly-charged pyridinium terminals. The absorption and fluorescence behavior of these dyes were compared to a commercially available fluorescent membrane stain, the styryl dye FM4-64. The hyperpolarizabilities of all dyes were compared using hyper-Rayleigh scattering at 800 nm. Cellular uptake, localization, toxicity and phototoxicity were evaluated using tissue cell cultures (HeLa, SK-OV-3 and MDA-231). Replacing the central alkene bridge of FM4-64 with a thiophene does not substantially change the absorption, fluorescence or hyperpolarizability, whereas changing the vinylene-links to ethynylenes shifts the absorption and fluorescence to shorter wavelengths, and reduces the hyperpolarizability by about a factor of two. SHG and fluorescence imaging experiments in live cells showed that the doubly-charged thiophene dyes localize in plasma membranes, and exhibit lower internalization rates compared to FM4-64, resulting in less signal from the cell cytosol. At a typical imaging concentration of 1 μM, the doubly-charged dyes showed no significant light or dark toxicity, whereas the singly-charged dyes are phototoxic even at 0.5 μM. The doubly-charged dyes showed phototoxicity at concentrations greater than 10 μM, although they do not generate singlet oxygen, indicating that the phototoxicity is type I rather than type II. The doubly-charged thiophene dyes are more effective than FM4-64 as SHG dyes for live cells.
Collapse
Affiliation(s)
- Ismael López-Duarte
- Department of Chemistry, Oxford University, Chemistry Research Laboratory, Oxford, UK OX1 3TA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Slater CR. The functional organization of motor nerve terminals. Prog Neurobiol 2015; 134:55-103. [DOI: 10.1016/j.pneurobio.2015.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/28/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
|
31
|
Hydrogen sulfide induces hyperpolarization and decreases the exocytosis of secretory granules of rat GH3 pituitary tumor cells. Biochem Biophys Res Commun 2015; 465:825-31. [DOI: 10.1016/j.bbrc.2015.08.095] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 01/05/2023]
|
32
|
Jonz MG, Zachar PC, Da Fonte DF, Mierzwa AS. Peripheral chemoreceptors in fish: A brief history and a look ahead. Comp Biochem Physiol A Mol Integr Physiol 2015; 186:27-38. [DOI: 10.1016/j.cbpa.2014.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 11/24/2022]
|
33
|
Balseiro-Gomez S, Flores JA, Acosta J, Ramirez-Ponce MP, Ales E. Identification of a New Exo-Endocytic Mechanism Triggered by Corticotropin-Releasing Hormone in Mast Cells. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26202981 DOI: 10.4049/jimmunol.1500253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The key role of mast cells (MC), either in development of inflammatory pathologies or in response to environmental stress, has been widely reported in recent years. Previous studies have described the effects of corticotropin-releasing hormone (CRH), which is released from inflamed tissues by cellular stress signals, on MC degranulation, a process possibly driven by selective secretion of mediators (piecemeal degranulation). In this study, we introduce a novel granular exo-endocytic pathway induced by CRH on peritoneal MC. We found that CRH triggers substantial exocytosis, which is even stronger than that induced by Ag stimulation and is characterized by large quantal size release events. Membrane fluorescence increases during stimulation in the presence of FM1-43 dye, corroborating the strength of this exocytosis, given that discrete upward fluorescence steps are often observed and suggesting that secretory granules are preferentially released by compound exocytosis. Additionally, the presence of a depot of large tubular organelles in the cytoplasm suggests that the exocytotic process is tightly coupled to a fast compound endocytosis. This CRH-stimulated mechanism is mediated through activation of adenylate cyclase and an increase of cAMP and intracellular Ca(2+), as evidenced by the fact that the effect of CRH is mimicked by forskolin and 8-bromo-cAMP. Thus, these outcomes constitute new evidence for the critical role of MC in pathophysiological conditions within a cellular stress environment and an alternative membrane trafficking route mediated by CRH.
Collapse
Affiliation(s)
- Santiago Balseiro-Gomez
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain
| | - Juan A Flores
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain
| | - Jorge Acosta
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain
| | - M Pilar Ramirez-Ponce
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain
| | - Eva Ales
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain
| |
Collapse
|
34
|
Gu H, Lazarenko RM, Koktysh D, Iacovitti L, Zhang Q. A Stem Cell-Derived Platform for Studying Single Synaptic Vesicles in Dopaminergic Synapses. Stem Cells Transl Med 2015; 4:887-93. [PMID: 26025981 DOI: 10.5966/sctm.2015-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/13/2015] [Indexed: 11/16/2022] Open
Abstract
The exocytotic release of dopamine is one of the most characteristic but also one of the least appreciated processes in dopaminergic neurotransmission. Fluorescence imaging has yielded rich information about the properties of synaptic vesicles and the release of neurotransmitters in excitatory and inhibitory neurons. In contrast, imaging-based studies for in-depth understanding of synaptic vesicle behavior in dopamine neurons are lagging largely because of a lack of suitable preparations. Midbrain culture has been one of the most valuable preparations for the subcellular investigation of dopaminergic transmission; however, the paucity and fragility of cultured dopaminergic neurons limits their use for live cell imaging. Recent developments in stem cell technology have led to the successful production of dopamine neurons from embryonic or induced pluripotent stem cells. Although the dopaminergic identity of these stem cell-derived neurons has been characterized in different ways, vesicle-mediated dopamine release from their axonal terminals has been barely assessed. We report a more efficient procedure to reliably generate dopamine neurons from embryonic stem cells, and it yields more dopamine neurons with more dopaminergic axon projections than midbrain culture does. Using a collection of functional measurements, we show that stem cell-derived dopamine neurons are indistinguishable from those in midbrain culture. Taking advantage of this new preparation, we simultaneously tracked the turnover of hundreds of synaptic vesicles individually using pH-sensitive quantum dots. By doing so, we revealed distinct fusion kinetics of the dopamine-secreting vesicles, which is consistent within both preparations.
Collapse
Affiliation(s)
- Haigang Gu
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Roman M Lazarenko
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Dmitry Koktysh
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Qi Zhang
- Department of Pharmacology and Vanderbilt Institute of Nanoscale Science and Engineering, Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
|
36
|
Flores JA, Balseiro-Gomez S, Cabeza JM, Acosta J, Ramirez-Ponce P, Ales E. A new role for myosin II in vesicle fission. PLoS One 2014; 9:e100757. [PMID: 24959909 PMCID: PMC4069105 DOI: 10.1371/journal.pone.0100757] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/28/2014] [Indexed: 11/19/2022] Open
Abstract
An endocytic vesicle is formed from a flat plasma membrane patch by a sequential process of invagination, bud formation and fission. The scission step requires the formation of a tubular membrane neck (the fission pore) that connects the endocytic vesicle with the plasma membrane. Progress in vesicle fission can be measured by the formation and closure of the fission pore. Live-cell imaging and sensitive biophysical measurements have provided various glimpses into the structure and behaviour of the fission pore. In the present study, the role of non-muscle myosin II (NM-2) in vesicle fission was tested by analyzing the kinetics of the fission pore with perforated-patch clamp capacitance measurements to detect single vesicle endocytosis with millisecond time resolution in peritoneal mast cells. Blebbistatin, a specific inhibitor of NM-2, dramatically increased the duration of the fission pore and also prevented closure during large endocytic events. Using the fluorescent markers FM1-43 and pHrodo Green dextran, we found that NM-2 inhibition greatly arrested vesicle fission in a late phase of the scission event when the pore reached a final diameter of ∼ 5 nm. Our results indicate that loss of the ATPase activity of myosin II drastically reduces the efficiency of membrane scission by making vesicle closure incomplete and suggest that NM-2 might be especially relevant in vesicle fission during compound endocytosis.
Collapse
Affiliation(s)
- Juan A. Flores
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Santiago Balseiro-Gomez
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Jose M. Cabeza
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Jorge Acosta
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Pilar Ramirez-Ponce
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Eva Ales
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
37
|
Monitoring of membrane phospholipid scrambling in human erythrocytes and K562 cells with FM1-43 - a comparison with annexin V-FITC. Cell Mol Biol Lett 2014; 19:262-76. [PMID: 24764144 PMCID: PMC6276018 DOI: 10.2478/s11658-014-0195-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 04/16/2014] [Indexed: 12/29/2022] Open
Abstract
The styryl dye FM1-43 becomes highly fluorescent upon binding to cell membranes. The breakdown of membrane phospholipid asymmetry in ionophore-stimulated T-lymphocytes further increases this fluorescence [Zweifach, 2000]. In this study, the capacity of FM1-43 to monitor membrane phospholipid scrambling was explored using flow cytometry in human erythrocytes and human erythrocyte progenitor K562 cells. The Ca2+-dependent phosphatidylserine-specific probe annexin V-FITC was used for comparison. The presented data show that the loss of phospholipid asymmetry that could be induced in human erythrocytes by elevated intracellular Ca2+ or by structurally different membrane intercalated amphiphilic compounds increases the FM1-43 fluorescence two- to fivefold. The profile of FM1-43 fluorescence for various treatments resembles that of phosphatidylserine exposure reported by annexin V-FITC. FM1-43 detected the onset of scrambling more efficiently than annexin V-FITC. The amphiphile-induced scrambling was shown to be a Ca2+-independent process. Monitoring of scrambling in K562 cells caused by NEM-induced Ca2+-release from intracellular stores and by Ca2+ and ionophore A23187 treatment showed that the increase in FM1-43 fluorescence correlated well with the number of annexin V-FITC-detected phosphatidylserine-positive cells. The results presented here show the usefulness of FM1-43 as a Ca2+-independent marker of dissipation in asymmetric membrane phospholipid distribution induced by various stimuli in both nucleated and non-nucleated cells.
Collapse
|
38
|
Castillo-Badillo JA, Cabrera-Wrooman A, García-Sáinz JA. Visualizing G protein-coupled receptors in action through confocal microscopy techniques. Arch Med Res 2014; 45:283-93. [PMID: 24751328 DOI: 10.1016/j.arcmed.2014.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 03/26/2014] [Indexed: 01/21/2023]
Abstract
G protein-coupled receptors constitute one of the most abundant entities in cellular communication. Elucidation of their structure and function as well as of their regulation began 30-40 years ago and the advance has markedly increased during the last 15 years. They participate in a plethora of cell functions such as regulation of metabolic fluxes, contraction, secretion, differentiation, or proliferation, and in essentially all activities of our organism; these receptors are targets of a large proportion of prescribed and illegal drugs. Fluorescence techniques have been used to study receptors for many years. The experimental result was usually a two-dimensional (2D) micrograph. Today, the result can be a spatiotemporal (four-dimensional, 4D) movie. Advances in microscopy, fluorescent protein design, and computer-assisted analysis have been of great importance to increase our knowledge on receptor regulation and function and create opportunities for future research. In this review we briefly depict the state of the art of the G protein-coupled receptor field and the methodologies used to study G protein-coupled receptor location, trafficking, dimerization, and other types of receptor-protein interaction. Fluorescence techniques now permit the capture of receptor images with high resolution and, together with a variety of fluorescent dyes that color organelles (such as the plasma membrane or the nucleus) or the cytoskeleton, allow researchers to obtain a much clearer idea of what is taking place at the cellular level. These developments are changing the way we explore cell communication and signal transduction, permitting deeper understanding of the physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Jean A Castillo-Badillo
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D.F., Mexico
| | | | - J Adolfo García-Sáinz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D.F., Mexico.
| |
Collapse
|
39
|
Fluorogenic probe for constitutive cellular endocytosis. ACTA ACUST UNITED AC 2013; 20:614-8. [PMID: 23601650 DOI: 10.1016/j.chembiol.2013.03.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/24/2013] [Accepted: 03/11/2013] [Indexed: 01/10/2023]
Abstract
Endocytosis is a fundamental process of eukaryotic cells that is critical for nutrient uptake, signal transduction, and growth. We have developed a molecular probe to quantify endocytosis. The probe is a lipid conjugated to a fluorophore that is masked with an enzyme-activatable moiety known as the trimethyl lock. The probe is not fluorescent when incorporated into the plasma membrane of human cells but becomes fluorescent upon internalization into endosomes, where cellular esterases activate the trimethyl lock. Using this probe, we found that human breast cancer cells undergo constitutive endocytosis more rapidly than do matched noncancerous cells. These data reveal a possible phenotypic distinction of cancer cells that could be the basis for chemotherapeutic intervention.
Collapse
|
40
|
Zheng LH, Wang CH, Shang SJ, Zhang XY, Wang YS, Wu QH, Hu MQ, Chai ZY, Wu X, Zheng H, Zhang C, Wang LC, Xiong W, Zhou Z. Real-time endocytosis imaging as a rapid assay of ligand-GPCR binding in single cells. Am J Physiol Cell Physiol 2013; 305:C751-60. [DOI: 10.1152/ajpcell.00335.2012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Most G protein-coupled receptors (GPCRs) do not generate membrane currents in response to ligand-receptor binding (LRB). Here, we describe a novel technique using endocytosis as a bioassay that can detect activation of a GPCR in a way analogous to patch-clamp recording of an ion channel in a living cell. The confocal imaging technique, termed FM endocytosis imaging (FEI), can record ligand-GPCR binding with high temporal (second) and spatial (micrometer) resolution. LRB leads to internalization of an endocytic vesicle, which can be labeled by a styryl FM dye and visualized as a fluorescent spot. Distinct from the green fluorescence protein-labeling method, FEI can detect LRB endocytosis mediated by essentially any receptors (GPCRs or receptors of tyrosine kinase) in a native cell/cell line. Three modified versions of FEI permit promising applications in functional GPCR studies and drug screening in living cells: 1) LRB can be recorded in “real time” (time scale of seconds); 2) internalized vesicles mediated by different GPCRs can be discriminated by different colors; and 3) a high throughput method can screen ligands of a specific GPCR.
Collapse
Affiliation(s)
- Liang-Hong Zheng
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chang-He Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Shu-Jiang Shang
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xiao-Yu Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Ye-Shi Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Qi-Hui Wu
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Mei-Qin Hu
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zu-Yin Chai
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xi Wu
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Hui Zheng
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chen Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Lie-Cheng Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Wei Xiong
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhuan Zhou
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
41
|
Rodrigues J, Silva RD, Noronha H, Pedras A, Gerós H, Côrte-Real M. Flow cytometry as a novel tool for structural and functional characterization of isolated yeast vacuoles. Microbiology (Reading) 2013; 159:848-856. [DOI: 10.1099/mic.0.062570-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Jorge Rodrigues
- Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Centro de Biologia Molecular e Ambiental (CBMA), Departamento de Biologia, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Rui D. Silva
- Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Centro de Biologia Molecular e Ambiental (CBMA), Departamento de Biologia, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Henrique Noronha
- Centro de Investigação e de Tecnologias Agro-Ambientais e Biológicas (CITAB), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Andreia Pedras
- Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Centro de Biologia Molecular e Ambiental (CBMA), Departamento de Biologia, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Hernâni Gerós
- Centro de Investigação e de Tecnologias Agro-Ambientais e Biológicas (CITAB), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Manuela Côrte-Real
- Departamento de Biologia, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Centro de Biologia Molecular e Ambiental (CBMA), Departamento de Biologia, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
42
|
Peng H, Kang N, Xu J, Stanton PK, Kang J. Two distinct modes of exocytotic fusion pore expansion in large astrocytic vesicles. J Biol Chem 2013; 288:16872-16881. [PMID: 23620588 DOI: 10.1074/jbc.m113.468231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Formation of the fusion pore is a central question for regulated exocytosis by which secretory cells release neurotransmitters or hormones. Here, by dynamically monitoring exocytosis of large vesicles (2-7 μM) in astrocytes with two-photon microscopy imaging, we found that the exocytotic fusion pore was generated from the SNARE-dependent fusion at a ring shape of the docked plasma-vesicular membrane and the movement of a fusion-produced membrane fragment. We observed two modes of fragment movements, 1) a shift fragment that shifted to expand the fusion pore and 2) a fall-in fragment that fell into the collapsed vesicle to expand the fusion pore. Shift and fall-in modes are associated with full and partial collapses of large vesicles, respectively. The astrocytic marker, sulforhodamine 101, stained the fusion-produced membrane fragment more brightly than FM 1-43. Sulforhodamine 101 imaging showed that double fusion pores could simultaneously occur in a single vesicle (16% of large vesicles) to accelerate discharge of vesicular contents. Electron microscopy of large astrocytic vesicles showed shift and fall-in membrane fragments. Two modes of fusion pore formation demonstrate a novel mechanism underlying fusion pore expansion and provide a new explanation for full and partial collapses of large secretory vesicles.
Collapse
Affiliation(s)
- Hong Peng
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595
| | - Ning Kang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595
| | - Jian Kang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595.
| |
Collapse
|
43
|
Properties of ribbon and non-ribbon release from rod photoreceptors revealed by visualizing individual synaptic vesicles. J Neurosci 2013; 33:2071-86. [PMID: 23365244 DOI: 10.1523/jneurosci.3426-12.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Vesicle release from rod photoreceptors is regulated by Ca(2+) entry through L-type channels located near synaptic ribbons. We characterized sites and kinetics of vesicle release in salamander rods by using total internal reflection fluorescence microscopy to visualize fusion of individual synaptic vesicles. A small number of vesicles were loaded by brief incubation with FM1-43 or a dextran-conjugated, pH-sensitive form of rhodamine, pHrodo. Labeled organelles matched the diffraction-limited size of fluorescent microspheres and disappeared rapidly during stimulation. Consistent with fusion, depolarization-evoked vesicle disappearance paralleled electrophysiological release kinetics and was blocked by inhibiting Ca(2+) influx. Rods maintained tonic release at resting membrane potentials near those in darkness, causing depletion of membrane-associated vesicles unless Ca(2+) entry was inhibited. This depletion of release sites implies that sustained release may be rate limited by vesicle delivery. During depolarizing stimulation, newly appearing vesicles approached the membrane at ∼800 nm/s, where they paused for ∼60 ms before fusion. With fusion, vesicles advanced ∼18 nm closer to the membrane. Release events were concentrated near ribbons, but lengthy depolarization also triggered release from more distant non-ribbon sites. Consistent with greater contributions from non-ribbon sites during lengthier depolarization, damaging the ribbon by fluorophore-assisted laser inactivation (FALI) of Ribeye caused only weak inhibition of exocytotic capacitance increases evoked by 200-ms depolarizing test steps, whereas FALI more strongly inhibited capacitance increases evoked by 25 ms steps. Amplifying release by use of non-ribbon sites when rods are depolarized in darkness may improve detection of decrements in release when they hyperpolarize to light.
Collapse
|
44
|
Abstract
Sustained neuronal communication relies on the coordinated activity of multiple proteins that regulate synaptic vesicle biogenesis and cycling within the presynaptic terminal. Synaptogyrin and synaptophysin are conserved MARVEL domain-containing transmembrane proteins that are among the most abundant synaptic vesicle constituents, although their role in the synaptic vesicle cycle has remained elusive. To further investigate the function of these proteins, we generated and characterized a synaptogyrin (gyr)-null mutant in Drosophila, whose genome encodes a single synaptogyrin isoform and lacks a synaptophysin homolog. We demonstrate that Drosophila synaptogyrin plays a modulatory role in synaptic vesicle biogenesis at larval neuromuscular junctions. Drosophila lacking synaptogyrin are viable and fertile and have no overt deficits in motor function. However, ultrastructural analysis of gyr larvae revealed increased synaptic vesicle diameter and enhanced variability in the size of synaptic vesicles. In addition, the resolution of endocytic cisternae into synaptic vesicles in response to strong stimulation is defective in gyr mutants. Electrophysiological analysis demonstrated an increase in quantal size and a concomitant decrease in quantal content, suggesting functional consequences for transmission caused by the loss of synaptogyrin. Furthermore, high-frequency stimulation resulted in increased facilitation and a delay in recovery from synaptic depression, indicating that synaptic vesicle exo-endocytosis is abnormally regulated during intense stimulation conditions. These results suggest that synaptogyrin modulates the synaptic vesicle exo-endocytic cycle and is required for the proper biogenesis of synaptic vesicles at nerve terminals.
Collapse
|
45
|
Automated condition-invariable neurite segmentation and synapse classification using textural analysis-based machine-learning algorithms. J Neurosci Methods 2012; 213:84-98. [PMID: 23261652 DOI: 10.1016/j.jneumeth.2012.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 11/24/2022]
Abstract
High-resolution live-cell imaging studies of neuronal structure and function are characterized by large variability in image acquisition conditions due to background and sample variations as well as low signal-to-noise ratio. The lack of automated image analysis tools that can be generalized for varying image acquisition conditions represents one of the main challenges in the field of biomedical image analysis. Specifically, segmentation of the axonal/dendritic arborizations in brightfield or fluorescence imaging studies is extremely labor-intensive and still performed mostly manually. Here we describe a fully automated machine-learning approach based on textural analysis algorithms for segmenting neuronal arborizations in high-resolution brightfield images of live cultured neurons. We compare performance of our algorithm to manual segmentation and show that it combines 90% accuracy, with similarly high levels of specificity and sensitivity. Moreover, the algorithm maintains high performance levels under a wide range of image acquisition conditions indicating that it is largely condition-invariable. We further describe an application of this algorithm to fully automated synapse localization and classification in fluorescence imaging studies based on synaptic activity. Textural analysis-based machine-learning approach thus offers a high performance condition-invariable tool for automated neurite segmentation.
Collapse
|
46
|
Cano R, Torres-Benito L, Tejero R, Biea AI, Ruiz R, Betz WJ, Tabares L. Structural and functional maturation of active zones in large synapses. Mol Neurobiol 2012; 47:209-19. [PMID: 22992975 DOI: 10.1007/s12035-012-8347-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/27/2012] [Indexed: 11/29/2022]
Abstract
Virtually all functions of the nervous system rely upon synapses, the sites of communication between neurons and between neurons and other cells. Synapses are complex structures, each one comprising hundreds of different types of molecules working in concert. They are organized by adhesive and scaffolding molecules that align presynaptic vesicular release sites, namely, active zones, with postsynaptic neurotransmitter receptors, thereby allowing rapid and reliable intercellular communication. Most synapses are relatively small, and acting alone exerts little effect on their postsynaptic partners. Some, however, are much larger and stronger, reliably driving the postsynaptic cell to its action potential threshold, acting essentially as electrical relays of excitation. These large synapses are among the best understood, and two of these are the subject of this review, namely, the vertebrate neuromuscular junction and the calyx of Held synapse in the mammalian auditory pathway of the brain stem. Both synapses undergo through a complex and well-coordinated maturation process, during which time the molecular elements and the biophysical properties of the secretory machinery are continuously adjusted to the synapse size and to the functional requirements. We here review the morphological and functional changes occurring during postnatal maturation, noting particular similarities and differences between these two large synapses.
Collapse
Affiliation(s)
- Raquel Cano
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, Avda. Sanchez Pizjuan 4, 41009, Seville, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
Fan J, Xia X, Li Y, Dy JG, Wong STC. A quantitative analytic pipeline for evaluating neuronal activities by high-throughput synaptic vesicle imaging. Neuroimage 2012; 62:2040-54. [PMID: 22732566 PMCID: PMC3437259 DOI: 10.1016/j.neuroimage.2012.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 06/12/2012] [Indexed: 11/26/2022] Open
Abstract
Synaptic vesicle dynamics play an important role in the study of neuronal and synaptic activities of neurodegradation diseases ranging from the epidemic Alzheimer's disease to the rare Rett syndrome. A high-throughput assay with a large population of neurons would be useful and efficient to characterize neuronal activity based on the dynamics of synaptic vesicles for the study of mechanisms or to discover drug candidates for neurodegenerative and neurodevelopmental disorders. However, the massive amounts of image data generated via high-throughput screening require enormous manual processing time and effort, restricting the practical use of such an assay. This paper presents an automated analytic system to process and interpret the huge data set generated by such assays. Our system enables the automated detection, segmentation, quantification, and measurement of neuron activities based on the synaptic vesicle assay. To overcome challenges such as noisy background, inhomogeneity, and tiny object size, we first employ MSVST (Multi-Scale Variance Stabilizing Transform) to obtain a denoised and enhanced map of the original image data. Then, we propose an adaptive thresholding strategy to solve the inhomogeneity issue, based on the local information, and to accurately segment synaptic vesicles. We design algorithms to address the issue of tiny objects of interest overlapping. Several post processing criteria are defined to filter false positives. A total of 152 features are extracted for each detected vesicle. A score is defined for each synaptic vesicle image to quantify the neuron activity. We also compare the unsupervised strategy with the supervised method. Our experiments on hippocampal neuron assays showed that the proposed system can automatically detect vesicles and quantify their dynamics for evaluating neuron activities. The availability of such an automated system will open opportunities for investigation of synaptic neuropathology and identification of candidate therapeutics for neurodegeneration.
Collapse
Affiliation(s)
- Jing Fan
- The Ting Tsung and Wei Fong Chao Center for Bioinformatics Research and Imaging for Neurosciences, The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
48
|
Togo T. Cell membrane disruption stimulates NO/PKG signaling and potentiates cell membrane repair in neighboring cells. PLoS One 2012; 7:e42885. [PMID: 22880128 PMCID: PMC3413670 DOI: 10.1371/journal.pone.0042885] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 07/12/2012] [Indexed: 02/05/2023] Open
Abstract
Resealing of a disrupted plasma membrane at the micron-diameter range requires Ca2+-regulated exocytosis. Repeated membrane disruptions reseal more quickly than the initial wound, and this potentiation of membrane resealing persists for at least 24 hours after the initial wound. Long-term potentiation of membrane resealing requires CREB-dependent gene expression, which is activated by the PKC- and p38 MAPK-dependent pathway in a wounded cell. The present study demonstrates that membrane resealing is potentiated in both wounded and neighboring cells in MDCK cells. Wounding of cells expressing CREB133, a mutant variant of CREB, does not show the potentiated response of cell membrane resealing in either wounded or neighboring cells. Furthermore, wounding of cells induces CREB phosphorylation, not only in wounded cells, but also in neighboring cells. Inhibition of the nitric oxide/PKG signaling pathway suppresses CREB phosphorylation in neighboring cells, but not in wounded cells. The potentiation of membrane resealing in neighboring cells is suppressed if the nitric oxide/PKG pathway is inhibited during the initial wound. Together, these results suggest that the nitric oxide/PKG pathway stimulates CREB phosphorylation in neighboring cells so that subsequent cell membrane disruptions of the neighboring cells reseal more quickly.
Collapse
Affiliation(s)
- Tatsuru Togo
- Department of Anatomy, St Marianna University School of Medicine, Kawasaki, Japan.
| |
Collapse
|
49
|
Daniel JA, Malladi CS, Kettle E, McCluskey A, Robinson PJ. Analysis of synaptic vesicle endocytosis in synaptosomes by high-content screening. Nat Protoc 2012; 7:1439-55. [PMID: 22767087 DOI: 10.1038/nprot.2012.070] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Small molecules modulating synaptic vesicle endocytosis (SVE) may ultimately be useful for diseases where pathological neurotransmission is implicated. Only a small number of specific SVE modulators have been identified to date. Slow progress is due to the laborious nature of traditional approaches to study SVE, in which nerve terminals are identified and studied in cultured neurons, typically yielding data from 10-20 synapses per experiment. We provide a protocol for a quantitative, high-throughput method for studying SVE in thousands of nerve terminals. Rat forebrain synaptosomes are attached to 96-well microplates and depolarized; SVE is then quantified by uptake of the dye FM4-64, which is imaged by high-content screening. Synaptosomes that have been frozen and stored can be used in place of fresh synaptosomes, reducing the experimental time and animal numbers required. With a supply of frozen synaptosomes, the assay can be performed within a day, including data analysis.
Collapse
Affiliation(s)
- James A Daniel
- Cell Signalling Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | | | | | | | | |
Collapse
|
50
|
Trueta C, Kuffler DP, De-Miguel FF. Cycling of dense core vesicles involved in somatic exocytosis of serotonin by leech neurons. Front Physiol 2012; 3:175. [PMID: 22685436 PMCID: PMC3368391 DOI: 10.3389/fphys.2012.00175] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 05/14/2012] [Indexed: 12/15/2022] Open
Abstract
We studied the cycling of dense core vesicles producing somatic exocytosis of serotonin. Our experiments were made using electron microscopy and vesicle staining with fluorescent dye FM1-43 in Retzius neurons of the leech, which secrete serotonin from clusters of dense core vesicles in a frequency-dependent manner. Electron micrographs of neurons at rest or after 1 Hz stimulation showed two pools of dense core vesicles. A perinuclear pool near Golgi apparatuses, from which vesicles apparently form, and a peripheral pool with vesicle clusters at a distance from the plasma membrane. By contrast, after 20 Hz electrical stimulation 47% of the vesicle clusters were apposed to the plasma membrane, with some omega exocytosis structures. Dense core and small clear vesicles apparently originating from endocytosis were incorporated in multivesicular bodies. In another series of experiments, neurons were stimulated at 20 Hz while bathed in a solution containing peroxidase. Electron micrographs of these neurons contained gold particles coupled to anti-peroxidase antibodies in dense core vesicles and multivesicular bodies located near the plasma membrane. Cultured neurons depolarized with high potassium in the presence of FM1-43 displayed superficial fluorescent spots, each reflecting a vesicle cluster. A partial bleaching of the spots followed by another depolarization in the presence of FM1-43 produced restaining of some spots, other spots disappeared, some remained without restaining and new spots were formed. Several hours after electrical stimulation the FM1-43 spots accumulated at the center of the somata. This correlated with electron micrographs of multivesicular bodies releasing their contents near Golgi apparatuses. Our results suggest that dense core vesicle cycling related to somatic serotonin release involves two steps: the production of clear vesicles and multivesicular bodies after exocytosis, and the formation of new dense core vesicles in the perinuclear region.
Collapse
Affiliation(s)
- Citlali Trueta
- Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz," México D. F., México
| | | | | |
Collapse
|