1
|
Heydecker M, Shitara A, Chen D, Tran DT, Masedunskas A, Tora MS, Ebrahim S, Appaduray MA, Galeano Niño JL, Bhardwaj A, Narayan K, Hardeman EC, Gunning PW, Weigert R. Coordination of force-generating actin-based modules stabilizes and remodels membranes in vivo. J Cell Biol 2024; 223:e202401091. [PMID: 39172125 PMCID: PMC11344176 DOI: 10.1083/jcb.202401091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Membrane remodeling drives a broad spectrum of cellular functions, and it is regulated through mechanical forces exerted on the membrane by cytoplasmic complexes. Here, we investigate how actin filaments dynamically tune their structure to control the active transfer of membranes between cellular compartments with distinct compositions and biophysical properties. Using intravital subcellular microscopy in live rodents we show that a lattice composed of linear filaments stabilizes the granule membrane after fusion with the plasma membrane and a network of branched filaments linked to the membranes by Ezrin, a regulator of membrane tension, initiates and drives to completion the integration step. Our results highlight how the actin cytoskeleton tunes its structure to adapt to dynamic changes in the biophysical properties of membranes.
Collapse
Affiliation(s)
- Marco Heydecker
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Akiko Shitara
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Pharmacology, Asahi University School of Dentistry, Gifu, Japan
| | - Desu Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Duy T Tran
- NIDCR Imaging Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Andrius Masedunskas
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Muhibullah S Tora
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mark A Appaduray
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Jorge Luis Galeano Niño
- EMBL Australia, Single Molecule Science node, University of New South Wales Sydney, Sydney, Australia
| | - Abhishek Bhardwaj
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Edna C Hardeman
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Peter W Gunning
- School of Biomedical Sciences, University of New South Wales Sydney, Sydney, Australia
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Koike S, Tachikawa M, Tsutsumi M, Okada T, Nemoto T, Keino-Masu K, Masu M. Actin dynamics switches two distinct modes of endosomal fusion in yolk sac visceral endoderm cells. eLife 2024; 13:RP95999. [PMID: 39441732 PMCID: PMC11498936 DOI: 10.7554/elife.95999] [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] [Indexed: 10/25/2024] Open
Abstract
Membranes undergo various patterns of deformation during vesicle fusion, but how this membrane deformation is regulated and contributes to fusion remains unknown. In this study, we developed a new method of observing the fusion of individual late endosomes and lysosomes by using mouse yolk sac visceral endoderm cells that have huge endocytic vesicles. We found that there were two distinct fusion modes that were differently regulated. In homotypic fusion, two late endosomes fused quickly, whereas in heterotypic fusion they fused to lysosomes slowly. Mathematical modeling showed that vesicle size is a critical determinant of these fusion types and that membrane fluctuation forces can overcome the vesicle size effects. We found that actin filaments were bound to late endosomes and forces derived from dynamic actin remodeling were necessary for quick fusion during homotypic fusion. Furthermore, cofilin played a role in endocytic fusion by regulating actin turnover. These data suggest that actin promotes vesicle fusion for efficient membrane trafficking in visceral endoderm cells.
Collapse
Affiliation(s)
- Seiichi Koike
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of ToyamaToyamaJapan
| | - Masashi Tachikawa
- Graduate School of Nanobioscience, Yokohama City UniversityYokohamaJapan
| | - Motosuke Tsutsumi
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
- National Institute for Physiological Sciences, National Institutes of Natural SciencesOkazakiJapan
| | - Takuya Okada
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| | - Tomomi Nemoto
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
- National Institute for Physiological Sciences, National Institutes of Natural SciencesOkazakiJapan
| | - Kazuko Keino-Masu
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| | - Masayuki Masu
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| |
Collapse
|
3
|
Heydecker M, Shitara A, Chen D, Tran D, Masedunskas A, Tora M, Ebrahim S, Appaduray MA, Galeano Niño JL, Bhardwaj A, Narayan K, Hardeman EC, Gunning PW, Weigert R. Spatial and Temporal Coordination of Force-generating Actin-based Modules Drives Membrane Remodeling In Vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.04.569944. [PMID: 38168275 PMCID: PMC10760165 DOI: 10.1101/2023.12.04.569944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Membrane remodeling drives a broad spectrum of cellular functions, and it is regulated through mechanical forces exerted on the membrane by cytoplasmic complexes. Here, we investigate how actin filaments dynamically tune their structure to control the active transfer of membranes between cellular compartments with distinct compositions and biophysical properties. Using intravital subcellular microscopy in live rodents we show that: a lattice composed of linear filaments stabilizes the granule membrane after fusion with the plasma membrane; and a network of branched filaments linked to the membranes by Ezrin, a regulator of membrane tension, initiates and drives to completion the integration step. Our results highlight how the actin cytoskeleton tunes its structure to adapt to dynamic changes in the biophysical properties of membranes.
Collapse
|
4
|
Biton T, Scher N, Carmon S, Elbaz-Alon Y, Schejter ED, Shilo BZ, Avinoam O. Fusion pore dynamics of large secretory vesicles define a distinct mechanism of exocytosis. J Cell Biol 2023; 222:e202302112. [PMID: 37707500 PMCID: PMC10501449 DOI: 10.1083/jcb.202302112] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/06/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Exocrine cells utilize large secretory vesicles (LSVs) up to 10 μm in diameter. LSVs fuse with the apical surface, often recruiting actomyosin to extrude their content through dynamic fusion pores. The molecular mechanism regulating pore dynamics remains largely uncharacterized. We observe that the fusion pores of LSVs in the Drosophila larval salivary glands expand, stabilize, and constrict. Arp2/3 is essential for pore expansion and stabilization, while myosin II is essential for pore constriction. We identify several Bin-Amphiphysin-Rvs (BAR) homology domain proteins that regulate fusion pore expansion and stabilization. We show that the I-BAR protein Missing-in-Metastasis (MIM) localizes to the fusion site and is essential for pore expansion and stabilization. The MIM I-BAR domain is essential but not sufficient for localization and function. We conclude that MIM acts in concert with actin, myosin II, and additional BAR-domain proteins to control fusion pore dynamics, mediating a distinct mode of exocytosis, which facilitates actomyosin-dependent content release that maintains apical membrane homeostasis during secretion.
Collapse
Affiliation(s)
- Tom Biton
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Scher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shari Carmon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Elbaz-Alon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D. Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
5
|
Ramezani A, Britton S, Zandi R, Alber M, Nematbakhsh A, Chen W. A multiscale chemical-mechanical model predicts impact of morphogen spreading on tissue growth. NPJ Syst Biol Appl 2023; 9:16. [PMID: 37210381 DOI: 10.1038/s41540-023-00278-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 05/03/2023] [Indexed: 05/22/2023] Open
Abstract
The exact mechanism controlling cell growth remains a grand challenge in developmental biology and regenerative medicine. The Drosophila wing disc tissue serves as an ideal biological model to study mechanisms involved in growth regulation. Most existing computational models for studying tissue growth focus specifically on either chemical signals or mechanical forces. Here we developed a multiscale chemical-mechanical model to investigate the growth regulation mechanism based on the dynamics of a morphogen gradient. By comparing the spatial distribution of dividing cells and the overall tissue shape obtained in model simulations with experimental data of the wing disc, it is shown that the size of the domain of the Dpp morphogen is critical in determining tissue size and shape. A larger tissue size with a faster growth rate and more symmetric shape can be achieved if the Dpp gradient spreads in a larger domain. Together with Dpp absorbance at the peripheral zone, the feedback regulation that downregulates Dpp receptors on the cell membrane allows for further spreading of the morphogen away from its source region, resulting in prolonged tissue growth at a more spatially homogeneous growth rate.
Collapse
Affiliation(s)
- Alireza Ramezani
- Department of Physics and Astronomy, University of California, Riverside, CA, 92521, USA
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
| | - Samuel Britton
- Department of Mathematics, University of California, Riverside, CA, 92521, USA
| | - Roya Zandi
- Department of Physics and Astronomy, University of California, Riverside, CA, 92521, USA
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
| | - Mark Alber
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
- Department of Mathematics, University of California, Riverside, CA, 92521, USA
| | - Ali Nematbakhsh
- Department of Mathematics, University of California, Riverside, CA, 92521, USA.
| | - Weitao Chen
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA.
- Department of Mathematics, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
6
|
Lehne F, Bogdan S. Swip-1 promotes exocytosis of glue granules in the exocrine Drosophila salivary gland. J Cell Sci 2023; 136:286884. [PMID: 36727484 PMCID: PMC10038153 DOI: 10.1242/jcs.260366] [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: 06/22/2022] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Exocytosis is a fundamental cellular process by which cells secrete cargos from their apical membrane into the extracellular lumen. Cargo release proceeds in sequential steps that depend on coordinated assembly and organization of an actin cytoskeletal network. Here, we identified the conserved actin-crosslinking protein Swip-1 as a novel regulator controlling exocytosis of glue granules in the Drosophila salivary gland. Real-time imaging revealed that Swip-1 is simultaneously recruited with F-actin onto secreting granules in proximity to the apical membrane. We observed that Swip-1 is rapidly cleared at the point of secretory vesicle fusion and colocalizes with actomyosin network around the fused vesicles. Loss of Swip-1 function impairs secretory cargo expulsion, resulting in strongly delayed secretion. Thus, our results uncover a novel role of Swip-1 in secretory vesicle compression and expulsion of cargo during regulated exocytosis. Remarkably, this function neither requires Ca2+ binding nor dimerization of Swip-1. Our data rather suggest that Swip-1 regulates actomyosin activity upstream of Rho-GTPase signaling to drive proper vesicle membrane crumpling and expulsion of cargo.
Collapse
Affiliation(s)
- Franziska Lehne
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Sven Bogdan
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, 35037 Marburg, Germany
| |
Collapse
|
7
|
Scheele CLGJ, Herrmann D, Yamashita E, Celso CL, Jenne CN, Oktay MH, Entenberg D, Friedl P, Weigert R, Meijboom FLB, Ishii M, Timpson P, van Rheenen J. Multiphoton intravital microscopy of rodents. NATURE REVIEWS. METHODS PRIMERS 2022; 2:89. [PMID: 37621948 PMCID: PMC10449057 DOI: 10.1038/s43586-022-00168-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 08/26/2023]
Abstract
Tissues are heterogeneous with respect to cellular and non-cellular components and in the dynamic interactions between these elements. To study the behaviour and fate of individual cells in these complex tissues, intravital microscopy (IVM) techniques such as multiphoton microscopy have been developed to visualize intact and live tissues at cellular and subcellular resolution. IVM experiments have revealed unique insights into the dynamic interplay between different cell types and their local environment, and how this drives morphogenesis and homeostasis of tissues, inflammation and immune responses, and the development of various diseases. This Primer introduces researchers to IVM technologies, with a focus on multiphoton microscopy of rodents, and discusses challenges, solutions and practical tips on how to perform IVM. To illustrate the unique potential of IVM, several examples of results are highlighted. Finally, we discuss data reproducibility and how to handle big imaging data sets.
Collapse
Affiliation(s)
- Colinda L. G. J. Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Department, Sydney, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Erika Yamashita
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Cristina Lo Celso
- Department of Life Sciences and Centre for Hematology, Imperial College London, London, UK
- Sir Francis Crick Institute, London, UK
| | - Craig N. Jenne
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Maja H. Oktay
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - David Entenberg
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Peter Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
- David H. Koch Center for Applied Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Franck L. B. Meijboom
- Department of Population Health Sciences, Sustainable Animal Stewardship, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Faculty of Humanities, Ethics Institute, Utrecht University, Utrecht, Netherlands
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Department, Sydney, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
8
|
Melis N, Subramanian B, Chen D, Weigert R. Imaging Neutrophil Migration in the Mouse Skin to Investigate Subcellular Membrane Remodeling Under Physiological Conditions. J Vis Exp 2022:10.3791/63581. [PMID: 35635466 PMCID: PMC10575475 DOI: 10.3791/63581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023] Open
Abstract
The study of immune cell recruitment and function in tissues has been a very active field over the last two decades. Neutrophils are among the first immune cells to reach the site of inflammation and to participate in the innate immune response during infection or tissue damage. So far, neutrophil migration has been successfully visualized using various in vitro experimental systems based on uniform stimulation, or confined migration under agarose, or micro-fluidic channels. However, these models do not recapitulate the complex microenvironment that neutrophils encounter in vivo. The development of multiphoton microscopy (MPM)-based techniques, such as intravital subcellular microscopy (ISMic), offer a unique tool to visualize and investigate neutrophil dynamics at subcellular resolutions under physiological conditions. In particular, the ear of a live anesthetized mouse provides an experimental advantage to follow neutrophil interstitial migration in real-time due to its ease of accessibility and lack of surgical exposure. ISMic provides the optical resolution, speed, and depth of acquisition necessary to track both cellular and, more importantly, subcellular processes in 3D over time (4D). Moreover, multi-modal imaging of the interstitial microenvironment (i.e., blood vessels, resident cells, extracellular matrix) can be readily accomplished using a combination of transgenic mice expressing select fluorescent markers, exogenous labeling via fluorescent probes, tissue intrinsic fluorescence, and second/third harmonic generated signals. This protocol describes 1) the preparation of neutrophils for adoptive transfer into the mouse ear, 2) different settings for optimal sub-cellular imaging, 3) strategies to minimize motion artifacts while maintaining a physiological response, 4) examples of membrane remodeling observed in neutrophils using ISMic, and 5) a workflow for the quantitative analysis of membrane remodeling in migrating neutrophils in vivo.
Collapse
Affiliation(s)
- Nicolas Melis
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute;
| | - Bhagawat Subramanian
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute; Lineberger Comprehensive Cancer Center, UNC-Chapel Hill
| | - Desu Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute;
| |
Collapse
|
9
|
Kamalesh K, Scher N, Biton T, Schejter ED, Shilo BZ, Avinoam O. Exocytosis by vesicle crumpling maintains apical membrane homeostasis during exocrine secretion. Dev Cell 2021; 56:1603-1616.e6. [PMID: 34102104 PMCID: PMC8191493 DOI: 10.1016/j.devcel.2021.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/17/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022]
Abstract
Exocrine secretion commonly employs micron-scale vesicles that fuse to a limited apical surface, presenting an extreme challenge for maintaining membrane homeostasis. Using Drosophila melanogaster larval salivary glands, we show that the membranes of fused vesicles undergo actomyosin-mediated folding and retention, which prevents them from incorporating into the apical surface. In addition, the diffusion of proteins and lipids between the fused vesicle and the apical surface is limited. Actomyosin contraction and membrane crumpling are essential for recruiting clathrin-mediated endocytosis to clear the retained vesicular membrane. Finally, we also observe membrane crumpling in secretory vesicles of the mouse exocrine pancreas. We conclude that membrane sequestration by crumpling followed by targeted endocytosis of the vesicular membrane, represents a general mechanism of exocytosis that maintains membrane homeostasis in exocrine tissues that employ large secretory vesicles.
Collapse
Affiliation(s)
- Kumari Kamalesh
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel; Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Scher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Tom Biton
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel; Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
Li D, Jia H, Zhang H, Lv M, Liu J, Zhang Y, Huang T, Huang B. TLR4 signaling induces the release of microparticles by tumor cells that regulate inflammatory cytokine IL-6 of macrophages via microRNA let-7b. Oncoimmunology 2021; 1:687-693. [PMID: 22934260 PMCID: PMC3429572 DOI: 10.4161/onci.19854] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tumor cells expressing TLRs is generally recognized to mediate tumor inflammation. However, whether and how tumor TLR signaling pathways negatively regulate tumor inflammation remains unclear. In this report, we find that TLR4 signaling of H22 hepatocarcinoma tumor cells is transduced through MyD88 pathway to actin cytoskeletons, leading to the release of microparticles (MPs), the cellular membrane-derived vesicles. As a result, tumor macrophages take up MPs and acquire MP-contained microRNA let-7b, which attenuates tumor inflammation by targeting proinflammatory cytokine IL-6. Thus, tumor TLR signaling, contrary to the original promoting effect, may play an opposite role in downregulating tumor inflammation through MP pathways.
Collapse
Affiliation(s)
- Dapeng Li
- Department of Biochemistry and Molecular Biology; Tongji Medical College; Huazhong University of Science and Technology; Wuhan, China ; Department of Breast and Thyroid Surgery; Union Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Erreni M, Doni A, Weigert R. Method for Acute Intravital Imaging of the Large Intestine in Live Mice. Methods Mol Biol 2021; 2304:285-299. [PMID: 34028723 DOI: 10.1007/978-1-0716-1402-0_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Intravital microscopy is an imaging technique aimed at the visualization of the dynamics of biological processes in live animals. In the last decade, the development of nonlinear optical microscopy has enormously increased the use of this technique, thus addressing key biological questions in different fields such as immunology, neurobiology and tumor biology. In addition, new upcoming strategies to minimize motion artifacts due to animal respiration and heartbeat have enabled the visualization in real time of biological processes at cellular and subcellular resolution. Recently, intravital microscopy has been applied to analyze different aspect of mucosal immunity in the gut. However, the majority of these studies have been performed on the small intestine. Although crucial aspects of the biology of this organ have been unveiled, the majority of intestinal pathologies in humans occur in the large intestine.Here, we describe a method to surgically expose and stabilize the large intestine in live mice and to perform short-term (up to 2 h) intravital microscopy.
Collapse
Affiliation(s)
- Marco Erreni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Andrea Doni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
12
|
Di Pasquale G, Perez Riveros P, Tora M, Sheikh T, Son A, Teos L, Grewe B, Swaim WD, Afione S, Zheng C, Jang SI, Shitara A, Alevizos I, Weigert R, Chiorini JA. Transduction of Salivary Gland Acinar Cells with a Novel AAV Vector 44.9. Mol Ther Methods Clin Dev 2020; 19:459-466. [PMID: 33294494 PMCID: PMC7689275 DOI: 10.1016/j.omtm.2020.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/09/2020] [Indexed: 11/20/2022]
Abstract
The loss of salivary gland function caused by radiation therapy of the head and neck or autoimmune disease such as Sjögren's syndrome is a serious condition that affects a patient's quality of life. Due to the combined exocrine and endocrine functions of the salivary gland, gene transfer to the salivary glands holds the potential for developing therapies for disorders of the salivary gland and the expression of therapeutic proteins via the exocrine pathway to the mouth, upper gastrointestinal tract, or endocrine pathway, systemically, into the blood. Recent clinical success with viral vector-mediated gene transfer for the treatment of irradiation-induced damage to the salivary glands has highlighted the need for the development of novel vectors with acinar cell tropism able to result in stable long-term transduction. Previous studies with adeno-associated virus (AAV) focused on the submandibular gland and reported mostly ductal cell transduction. In this study, we have screened AAV vectors for acinar cell tropism in the parotid gland utilizing membrane-tomato floxed membrane-GFP transgenic mice to screen CRE recombinase encoding AAV vectors of different clades to rapidly identify capsid isolates able to transduce salivary gland acinar cells. We determined that AAVRh10 and a novel isolate found as a contaminant of a laboratory stock of simian adenovirus SV15, AAV44.9, are both able to transduce parotid and sublingual acinar cells. Persistence and localization of transduction of these AAVs were tested using vectors encoding firefly luciferase, which was detected 6 months after vector administration. Most luciferase expression was localized to the salivary gland compared to that of distal organs. Transduction resulted in robust secretion of recombinant protein in both blood and saliva. Transduction was species specific, with AAVRh10 having stronger transduction activity in rats compared with AAV44.9 or AAV2 but weaker in human primary salivary gland cells. This work demonstrates efficient transduction of parotid acinar cells by AAV that resulted in secretion of recombinant protein in both serum and saliva.
Collapse
Affiliation(s)
- Giovanni Di Pasquale
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paola Perez Riveros
- Salivary Gland Biology and Disorder Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Muhibullah Tora
- Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tayyab Sheikh
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aran Son
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Leyla Teos
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brigitte Grewe
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - William D. Swaim
- Salivary Gland Biology and Disorder Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra Afione
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Changyu Zheng
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shyh-Ing Jang
- Salivary Gland Biology and Disorder Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Akiko Shitara
- Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ilias Alevizos
- Sjögren’s Syndrome and Salivary Gland Dysfunction Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Roberto Weigert
- Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - John A. Chiorini
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Loganathan R, Kim JH, Wells MB, Andrew DJ. Secrets of secretion-How studies of the Drosophila salivary gland have informed our understanding of the cellular networks underlying secretory organ form and function. Curr Top Dev Biol 2020; 143:1-36. [PMID: 33820619 DOI: 10.1016/bs.ctdb.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Secretory organs are critical for organismal survival. Yet, the transcriptional regulatory mechanisms governing their development and maintenance remain unclear for most model secretory organs. The Drosophila embryonic salivary gland (SG) remedies this deficiency as one of the few organs wherein direct connections from the expression of the early patterning genes to cell specification to organ architecture and functional specialization can be made. Few other models of secretion can be accorded this distinction. Studies from the past three decades have made enormous strides in parsing out the roles of distinct transcription factors (TFs) that direct major steps in furnishing this secretory organ. In the first step of specifying the salivary gland, the activity of the Hox factors Sex combs reduced, Extradenticle, and Homothorax activate expression of fork head (fkh), sage, and CrebA, which code for the major suite of TFs that carry forward the task of organ building and maintenance. Then, in the second key step of building the SG, the program for cell fate maintenance and morphogenesis is deployed. Fkh maintains the secretory cell fate by regulating its own expression and that of sage and CrebA. Fkh and Sage maintain secretory cell viability by actively blocking apoptotic cell death. Fkh, along with two other TFs, Hkb and Rib, also coordinates organ morphogenesis, transforming two plates of precursor cells on the embryo surface into elongated internalized epithelial tubes. Acquisition of functional specialization, the third key step, is mediated by CrebA and Fkh working in concert with Sage and yet another TF, Sens. CrebA directly upregulates expression of all of the components of the secretory machinery as well as other genes (e.g., Xbp1) necessary for managing the physiological stress that inexorably accompanies high secretory load. Secretory cargo specificity is controlled by Sage and Sens in collaboration with Fkh. Investigations have also uncovered roles for various signaling pathways, e.g., Dpp signaling, EGF signaling, GPCR signaling, and cytoskeletal signaling, and their interactions within the gene regulatory networks that specify, build, and specialize the SG. Collectively, studies of the SG have expanded our knowledge of secretory dynamics, cell polarity, and cytoskeletal mechanics in the context of organ development and function. Notably, the embryonic SG has made the singular contribution as a model system that revealed the core function of CrebA in scaling up secretory capacity, thus, serving as the pioneer system in which the conserved roles of the mammalian Creb3/3L-family orthologues were first discovered.
Collapse
Affiliation(s)
- Rajprasad Loganathan
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ji Hoon Kim
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael B Wells
- Idaho College of Osteopathic Medicine, Meridian, ID, United States
| | - Deborah J Andrew
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
14
|
Miklavc P, Frick M. Actin and Myosin in Non-Neuronal Exocytosis. Cells 2020; 9:cells9061455. [PMID: 32545391 PMCID: PMC7348895 DOI: 10.3390/cells9061455] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/18/2022] Open
Abstract
Cellular secretion depends on exocytosis of secretory vesicles and discharge of vesicle contents. Actin and myosin are essential for pre-fusion and post-fusion stages of exocytosis. Secretory vesicles depend on actin for transport to and attachment at the cell cortex during the pre-fusion phase. Actin coats on fused vesicles contribute to stabilization of large vesicles, active vesicle contraction and/or retrieval of excess membrane during the post-fusion phase. Myosin molecular motors complement the role of actin. Myosin V is required for vesicle trafficking and attachment to cortical actin. Myosin I and II members engage in local remodeling of cortical actin to allow vesicles to get access to the plasma membrane for membrane fusion. Myosins stabilize open fusion pores and contribute to anchoring and contraction of actin coats to facilitate vesicle content release. Actin and myosin function in secretion is regulated by a plethora of interacting regulatory lipids and proteins. Some of these processes have been first described in non-neuronal cells and reflect adaptations to exocytosis of large secretory vesicles and/or secretion of bulky vesicle cargoes. Here we collate the current knowledge and highlight the role of actomyosin during distinct phases of exocytosis in an attempt to identify unifying molecular mechanisms in non-neuronal secretory cells.
Collapse
Affiliation(s)
- Pika Miklavc
- School of Science, Engineering & Environment, University of Salford, Manchester M5 4WT, UK
- Correspondence: (P.M.); (M.F.); Tel.: +44-0161-295-3395 (P.M.); +49-731-500-23115 (M.F.); Fax: +49-731-500-23242 (M.F.)
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Correspondence: (P.M.); (M.F.); Tel.: +44-0161-295-3395 (P.M.); +49-731-500-23115 (M.F.); Fax: +49-731-500-23242 (M.F.)
| |
Collapse
|
15
|
Müller MT, Schempp R, Lutz A, Felder T, Felder E, Miklavc P. Interaction of microtubules and actin during the post-fusion phase of exocytosis. Sci Rep 2019; 9:11973. [PMID: 31427591 PMCID: PMC6700138 DOI: 10.1038/s41598-019-47741-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/09/2019] [Indexed: 01/24/2023] Open
Abstract
Exocytosis is the intracellular trafficking step where a secretory vesicle fuses with the plasma membrane to release vesicle content. Actin and microtubules both play a role in exocytosis; however, their interplay is not understood. Here we study the interaction of actin and microtubules during exocytosis in lung alveolar type II (ATII) cells that secrete surfactant from large secretory vesicles. Surfactant extrusion is facilitated by an actin coat that forms on the vesicle shortly after fusion pore opening. Actin coat compression allows hydrophobic surfactant to be released from the vesicle. We show that microtubules are localized close to actin coats and stay close to the coats during their compression. Inhibition of microtubule polymerization by colchicine and nocodazole affected the kinetics of actin coat formation and the extent of actin polymerisation on fused vesicles. In addition, microtubule and actin cross-linking protein IQGAP1 localized to fused secretory vesicles and IQGAP1 silencing influenced actin polymerisation after vesicle fusion. This study demonstrates that microtubules can influence actin coat formation and actin polymerization on secretory vesicles during exocytosis.
Collapse
Affiliation(s)
- M Tabitha Müller
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Rebekka Schempp
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Anngrit Lutz
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Tatiana Felder
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Edward Felder
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Pika Miklavc
- School of Environment and Life Sciences, University of Salford, The Crescent, M54WT, Salford, United Kingdom.
| |
Collapse
|
16
|
Ebrahim S, Weigert R. Intravital microscopy in mammalian multicellular organisms. Curr Opin Cell Biol 2019; 59:97-103. [PMID: 31125832 PMCID: PMC6726551 DOI: 10.1016/j.ceb.2019.03.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/25/2019] [Accepted: 03/29/2019] [Indexed: 12/22/2022]
Abstract
Imaging subcellular processes in live animals is no longer a dream. The development of Intravital Subcellular Microscopy (ISMic) combined with the astounding repertoire of available mouse models makes it possible to investigate processes such as membrane trafficking in mammalian living tissues under native conditions. This has provided the unique opportunity to answer questions that cannot be otherwise addressed in reductionist model systems and to link cell biology to tissue pathophysiology.
Collapse
Affiliation(s)
- Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr. Rm 2050B, Bethesda, MD, 20892, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr. Rm 2050B, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Dynamic polyhedral actomyosin lattices remodel micron-scale curved membranes during exocytosis in live mice. Nat Cell Biol 2019; 21:933-939. [PMID: 31358965 DOI: 10.1038/s41556-019-0365-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/25/2019] [Indexed: 01/10/2023]
Abstract
Actomyosin networks, the cell's major force production machineries, remodel cellular membranes during myriad dynamic processes1,2 by assembling into various architectures with distinct force generation properties3,4. While linear and branched actomyosin architectures are well characterized in cell-culture and cell-free systems3, it is not known how actin and myosin networks form and function to remodel membranes in complex three-dimensional mammalian tissues. Here, we use four-dimensional spinning-disc confocal microscopy with image deconvolution to acquire macromolecular-scale detail of dynamic actomyosin networks in exocrine glands of live mice. We address how actin and myosin organize around large membrane-bound secretory vesicles and generate the forces required to complete exocytosis5-7. We find that actin and non-muscle myosin II (NMII) assemble into previously undescribed polyhedral-like lattices around the vesicle membrane. The NMII lattice comprises bipolar minifilaments8-10 as well as non-canonical three-legged configurations. Using photobleaching and pharmacological perturbations in vivo, we show that actomyosin contractility and actin polymerization together push on the underlying vesicle membrane to overcome the energy barrier and complete exocytosis7. Our imaging approach thus unveils a force-generating actomyosin lattice that regulates secretion in the exocrine organs of live animals.
Collapse
|
18
|
Samiei M, Ahmadian E, Eftekhari A, Eghbal MA, Rezaie F, Vinken M. Cell junctions and oral health. EXCLI JOURNAL 2019; 18:317-330. [PMID: 31338005 PMCID: PMC6635732 DOI: 10.17179/excli2019-1370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022]
Abstract
The oral cavity and its appendices are exposed to considerable environmental and mechanical stress. Cell junctions play a pivotal role in this context. Among those, gap junctions permit the exchange of compounds between cells, thereby controlling processes such as cell growth and differentiation. Tight junctions restrict paracellular transportation and inhibit movement of integral membrane proteins between the different plasma membrane poles. Adherens junctions attach cells one to another and provide a solid backbone for resisting to mechanistical stress. The integrity of oral mucosa, normal tooth development and saliva secretion depend on the proper function of all these types of cell junctions. Furthermore, deregulation of junctional proteins and/or mutations in their genes can alter tissue functioning and may result in various human disorders, including dental and periodontal problems, salivary gland malfunction, hereditary and infectious diseases as well as tumorigenesis. The present manuscript reviews the role of cell junctions in the (patho)physiology of the oral cavity and its appendices, including salivary glands.
Collapse
Affiliation(s)
- Mohammad Samiei
- Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Dental and Periodontal Research center, Tabriz University of Medical Sciences, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Pharmacology and Toxicology department, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mohammad Ali Eghbal
- Drug Applied Research Center and Pharmacology and Toxicology department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshte Rezaie
- General Practitioner, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
19
|
Patel S, Kim J, Herrera M, Mukherjee A, Kabanov AV, Sahay G. Brief update on endocytosis of nanomedicines. Adv Drug Deliv Rev 2019; 144:90-111. [PMID: 31419450 PMCID: PMC6986687 DOI: 10.1016/j.addr.2019.08.004] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/06/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022]
Abstract
The complexity of nanoscale interactions between biomaterials and cells has limited the realization of the ultimate vision of nanotechnology in diagnostics and therapeutics. As such, significant effort has been devoted to advancing our understanding of the biophysical interactions of the myriad nanoparticles. Endocytosis of nanomedicine has drawn tremendous interest in the last decade. Here, we highlight the ever-present barriers to efficient intracellular delivery of nanoparticles as well as the current advances and strategies deployed to breach these barriers. We also introduce new barriers that have been largely overlooked such as the glycocalyx and macromolecular crowding. Additionally, we draw attention to the potential complications arising from the disruption of the newly discovered functions of the lysosomes. Novel strategies of exploiting the inherent intracellular defects in disease states to enhance delivery and the use of exosomes for bioanalytics and drug delivery are explored. Furthermore, we discuss the advances in imaging techniques like electron microscopy, super resolution fluorescence microscopy, and single particle tracking which have been instrumental in our growing understanding of intracellular pathways and nanoparticle trafficking. Finally, we advocate for the push towards more intravital analysis of nanoparticle transport phenomena using the multitude of techniques available to us. Unraveling the underlying mechanisms governing the cellular barriers to delivery and biological interactions of nanoparticles will guide the innovations capable of breaching these barriers.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Marco Herrera
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Anindit Mukherjee
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA; Department of Biomedical Engineering, Oregon Health and Science University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA.
| |
Collapse
|
20
|
Klein O, Sagi-Eisenberg R. Anaphylactic Degranulation of Mast Cells: Focus on Compound Exocytosis. J Immunol Res 2019; 2019:9542656. [PMID: 31011586 PMCID: PMC6442490 DOI: 10.1155/2019/9542656] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 12/26/2018] [Indexed: 01/15/2023] Open
Abstract
Anaphylaxis is a notorious type 2 immune response which may result in a systemic response and lead to death. A precondition for the unfolding of the anaphylactic shock is the secretion of inflammatory mediators from mast cells in response to an allergen, mostly through activation of the cells via the IgE-dependent pathway. While mast cells are specialized secretory cells that can secrete through a variety of exocytic modes, the most predominant mode exerted by the mast cell during anaphylaxis is compound exocytosis-a specialized form of regulated exocytosis where secretory granules fuse to one another. Here, we review the modes of regulated exocytosis in the mast cell and focus on compound exocytosis. We review historical landmarks in the research of compound exocytosis in mast cells and the methods available for investigating compound exocytosis. We also review the molecular mechanisms reported to underlie compound exocytosis in mast cells and expand further with reviewing key findings from other cell types. Finally, we discuss the possible reasons for the mast cell to utilize compound exocytosis during anaphylaxis, the conflicting evidence in different mast cell models, and the open questions in the field which remain to be answered.
Collapse
Affiliation(s)
- Ofir Klein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Sagi-Eisenberg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
21
|
Porat-Shliom N, Harding OJ, Malec L, Narayan K, Weigert R. Mitochondrial Populations Exhibit Differential Dynamic Responses to Increased Energy Demand during Exocytosis In Vivo. iScience 2019; 11:440-449. [PMID: 30661001 PMCID: PMC6355620 DOI: 10.1016/j.isci.2018.12.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/14/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022] Open
Abstract
Mitochondria are dynamic organelles undergoing fission, fusion, and translocation. These processes have been studied in cultured cells; however, little is known about their regulation in cells within tissues in vivo. We applied four-dimensional intravital microscopy to address this in secretory cells of the salivary gland. We found that mitochondria are organized in two populations: one juxtaposed to the basolateral plasma membrane and the other dispersed in the cytosol. Under basal conditions, central mitochondria exhibit microtubule-dependent motility and low fusion rate, whereas basolateral mitochondria are static and display high fusion rate. Increasing cellular energy demand by β-adrenergic stimulation of regulated exocytosis selectively enhanced motility and fusion of central mitochondria. Inhibition of microtubule polymerization led to inhibition of central mitochondrial motility and fusion and a marked reduction in exocytosis. This study reveals a conserved heterogeneity in mitochondrial positioning and dynamics in exocrine tissues that may have fundamental implications in organ pathophysiology. In the salivary glands, mitochondria exist in two populations: basolateral and central Basolateral mitochondria are static and frequently fuse Central mitochondria are highly motile and rarely fuse Exocytosis elicits selective, microtubule-dependent response in central mitochondria
Collapse
Affiliation(s)
- Natalie Porat-Shliom
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; Cell Biology and Imaging Section, Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Olivia J Harding
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lenka Malec
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 8560 Progress Drive, Frederick, MD 21701, USA; Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
22
|
Shitara A, Malec L, Ebrahim S, Chen D, Bleck C, Hoffman MP, Weigert R. Cdc42 negatively regulates endocytosis during apical membrane maintenance in live animals. Mol Biol Cell 2018; 30:324-332. [PMID: 30540520 PMCID: PMC6589572 DOI: 10.1091/mbc.e18-10-0615] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Lumen establishment and maintenance are fundamental for tubular organs physiological functions. Most of the studies investigating the mechanisms regulating this process have been carried out in cell cultures or in smaller organisms, whereas little has been done in mammalian model systems in vivo. Here we used the salivary glands of live mice to examine the role of the small GTPase Cdc42 in the regulation of the homeostasis of the intercellular canaliculi, a specialized apical domain of the acinar cells, where protein and fluid secretion occur. Depletion of Cdc42 in adult mice induced a significant expansion of the apical canaliculi, whereas depletion at late embryonic stages resulted in a complete inhibition of their postnatal formation. In addition, intravital subcellular microscopy revealed that reduced levels of Cdc42 affected membrane trafficking from and toward the plasma membrane, highlighting a novel role for Cdc42 in membrane remodeling through the negative regulation of selected endocytic pathways.
Collapse
Affiliation(s)
- Akiko Shitara
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Lenka Malec
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Desu Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892.,College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD 20742
| | - Christopher Bleck
- Electron Microscopy Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institutes of Health, Bethesda, MD 20892
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892.,Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
23
|
Wong WY, Pier M, Limesand KH. Persistent disruption of lateral junctional complexes and actin cytoskeleton in parotid salivary glands following radiation treatment. Am J Physiol Regul Integr Comp Physiol 2018; 315:R656-R667. [PMID: 29897817 PMCID: PMC6230885 DOI: 10.1152/ajpregu.00388.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 02/02/2023]
Abstract
Xerostomia and hyposalivation are debilitating side effects for patients treated with ionizing radiation for head and neck cancer. Despite technological advances, collateral damage to the salivary glands remains a significant problem for patients and severely diminishes their quality of life. During the wound healing process, restoration of junctional contacts is necessary to maintain polarity, structural integrity, and orientation cues for secretion. However, little is known about whether these structural molecules are impacted following radiation damage and more importantly, during tissue restoration. We evaluated changes in adherens junctions and cytoskeletal regulators in an injury model where mice were irradiated with 5 Gy and a restoration model where mice injected postradiation with insulin-like growth factor 1 (IGF1) are capable of restoring salivary function. Using coimmunoprecipitation, there is a decrease in epithelial (E)-cadherin bound to β-catenin following damage that is restored to untreated levels with IGF1. Via its adaptor proteins, β-catenin links the cadherins to the cytoskeleton and part of this regulation is mediated through Rho-associated coiled-coil containing kinase (ROCK) signaling. In our radiation model, filamentous (F)-actin organization is fragmented, and there is an induction of ROCK activity. However, a ROCK inhibitor, Y-27632, prevents E-cadherin/β-catenin dissociation following radiation treatment. These findings illustrate that radiation induces a ROCK-dependent disruption of the cadherin-catenin complex and alters F-actin organization at stages of damage when hyposalivation is observed. Understanding the regulation of these components will be critical in the discovery of therapeutics that have the potential to restore function in polarized epithelium.
Collapse
Affiliation(s)
- Wen Yu Wong
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona , Tucson, Arizona
| | - Maricela Pier
- Department of Nutritional Sciences, University of Arizona , Tucson, Arizona
| | - Kirsten H Limesand
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona , Tucson, Arizona
- Department of Nutritional Sciences, University of Arizona , Tucson, Arizona
| |
Collapse
|
24
|
Ebrahim S, Liu J, Weigert R. The Actomyosin Cytoskeleton Drives Micron-Scale Membrane Remodeling In Vivo Via the Generation of Mechanical Forces to Balance Membrane Tension Gradients. Bioessays 2018; 40:e1800032. [PMID: 30080263 DOI: 10.1002/bies.201800032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/29/2018] [Indexed: 12/31/2022]
Abstract
The remodeling of biological membranes is crucial for a vast number of cellular activities and is an inherently multiscale process in both time and space. Seminal work has provided important insights into nanometer-scale membrane deformations, and highlighted the remarkable variation and complexity in the underlying molecular machineries and mechanisms. However, how membranes are remodeled at the micron-scale, particularly in vivo, remains poorly understood. Here, we discuss how using regulated exocytosis of large (1.5-2.0 μm) membrane-bound secretory granules in the salivary gland of live mice as a model system, has provided evidence for the importance of the actomyosin cytoskeleton in micron-scale membrane remodeling in physiological conditions. We highlight some of these advances, and present mechanistic hypotheses for how the various biochemical and biophysical properties of distinct actomyosin networks may drive this process.
Collapse
Affiliation(s)
- Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.,National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Liu
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Neumann NM, Perrone MC, Veldhuis JH, Huebner RJ, Zhan H, Devreotes PN, Brodland GW, Ewald AJ. Coordination of Receptor Tyrosine Kinase Signaling and Interfacial Tension Dynamics Drives Radial Intercalation and Tube Elongation. Dev Cell 2018; 45:67-82.e6. [PMID: 29634937 DOI: 10.1016/j.devcel.2018.03.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/11/2018] [Accepted: 03/14/2018] [Indexed: 11/16/2022]
Abstract
We sought to understand how cells collectively elongate epithelial tubes. We first used 3D culture and biosensor imaging to demonstrate that epithelial cells enrich Ras activity, phosphatidylinositol (3,4,5)-trisphosphate (PIP3), and F-actin to their leading edges during migration within tissues. PIP3 enrichment coincided with, and could enrich despite inhibition of, F-actin dynamics, revealing a conserved migratory logic compared with single cells. We discovered that migratory cells can intercalate into the basal tissue surface and contribute to tube elongation. We then connected molecular activities to subcellular mechanics using force inference analysis. Migration and transient intercalation required specific and similar anterior-posterior ratios of interfacial tension. Permanent intercalations were distinguished by their capture at the boundary through time-varying tension dynamics. Finally, we integrated our experimental and computational data to generate a finite element model of tube elongation. Our model revealed that intercalation, interfacial tension dynamics, and high basal stress are together sufficient for mammary morphogenesis.
Collapse
Affiliation(s)
- Neil M Neumann
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Rangos 452, Baltimore, MD 21205, USA
| | - Matthew C Perrone
- Department of Civil and Environmental Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Jim H Veldhuis
- Department of Civil and Environmental Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Robert J Huebner
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Rangos 452, Baltimore, MD 21205, USA
| | - Huiwang Zhan
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Rangos 452, Baltimore, MD 21205, USA
| | - Peter N Devreotes
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Rangos 452, Baltimore, MD 21205, USA
| | - G Wayne Brodland
- Department of Civil and Environmental Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada; Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Andrew J Ewald
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Rangos 452, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Ficht X, Thelen F, Stolp B, Stein JV. Preparation of Murine Submandibular Salivary Gland for Upright Intravital Microscopy. J Vis Exp 2018. [PMID: 29781999 DOI: 10.3791/57283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The submandibular salivary gland (SMG) is one of the three major salivary glands, and is of interest for many different fields of biological research, including cell biology, oncology, dentistry, and immunology. The SMG is an exocrine gland comprised of secretory epithelial cells, myofibroblasts, endothelial cells, nerves, and extracellular matrix. Dynamic cellular processes in the rat and mouse SMG have previously been imaged, mostly using inverted multi-photon microscope systems. Here, we describe a straightforward protocol for the surgical preparation and stabilization of the murine SMG in anesthetized mice for in vivo imaging with upright multi-photon microscope systems. We present representative intravital image sets of endogenous and adoptively transferred fluorescent cells, including the labeling of blood vessels or salivary ducts and second harmonic generation to visualize fibrillar collagen. In sum, our protocol allows for surgical preparation of mouse salivary glands in upright microscopy systems, which are commonly used for intravital imaging in the field of immunology.
Collapse
Affiliation(s)
- Xenia Ficht
- Theodor-Kocher Institute, University of Bern
| | | | - Bettina Stolp
- Theodor-Kocher Institute, University of Bern; Center for Infectious Diseases, Integrative Virology, University Clinic of Heidelberg
| | | |
Collapse
|
27
|
Masedunskas A, Appaduray MA, Lucas CA, Lastra Cagigas M, Heydecker M, Holliday M, Meiring JCM, Hook J, Kee A, White M, Thomas P, Zhang Y, Adelstein RS, Meckel T, Böcking T, Weigert R, Bryce NS, Gunning PW, Hardeman EC. Parallel assembly of actin and tropomyosin, but not myosin II, during de novo actin filament formation in live mice. J Cell Sci 2018; 131:jcs.212654. [PMID: 29487177 DOI: 10.1242/jcs.212654] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/12/2018] [Indexed: 01/04/2023] Open
Abstract
Many actin filaments in animal cells are co-polymers of actin and tropomyosin. In many cases, non-muscle myosin II associates with these co-polymers to establish a contractile network. However, the temporal relationship of these three proteins in the de novo assembly of actin filaments is not known. Intravital subcellular microscopy of secretory granule exocytosis allows the visualisation and quantification of the formation of an actin scaffold in real time, with the added advantage that it occurs in a living mammal under physiological conditions. We used this model system to investigate the de novo assembly of actin, tropomyosin Tpm3.1 (a short isoform of TPM3) and myosin IIA (the form of non-muscle myosin II with its heavy chain encoded by Myh9) on secretory granules in mouse salivary glands. Blocking actin polymerization with cytochalasin D revealed that Tpm3.1 assembly is dependent on actin assembly. We used time-lapse imaging to determine the timing of the appearance of the actin filament reporter LifeAct-RFP and of Tpm3.1-mNeonGreen on secretory granules in LifeAct-RFP transgenic, Tpm3.1-mNeonGreen and myosin IIA-GFP (GFP-tagged MYH9) knock-in mice. Our findings are consistent with the addition of tropomyosin to actin filaments shortly after the initiation of actin filament nucleation, followed by myosin IIA recruitment.
Collapse
Affiliation(s)
| | | | | | | | - Marco Heydecker
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia.,Membrane Dynamics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287 Darmstadt, Germany
| | - Mira Holliday
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| | | | - Jeff Hook
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| | - Anthony Kee
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| | - Melissa White
- South Australian Genome Editing, Facility Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Paul Thomas
- South Australian Genome Editing, Facility Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Yingfan Zhang
- NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Tobias Meckel
- Membrane Dynamics, Department of Biology, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287 Darmstadt, Germany
| | - Till Böcking
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, CCR, National Cancer Institute, Bethesda, MD 20892, USA
| | - Nicole S Bryce
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| | - Peter W Gunning
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| | - Edna C Hardeman
- School of Medical Sciences, UNSW Sydney, NSW 2052, Australia
| |
Collapse
|
28
|
Kee AJ, Chagan J, Chan JY, Bryce NS, Lucas CA, Zeng J, Hook J, Treutlein H, Laybutt DR, Stehn JR, Gunning PW, Hardeman EC. On-target action of anti-tropomyosin drugs regulates glucose metabolism. Sci Rep 2018; 8:4604. [PMID: 29545590 PMCID: PMC5854615 DOI: 10.1038/s41598-018-22946-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/01/2018] [Indexed: 01/09/2023] Open
Abstract
The development of novel small molecule inhibitors of the cancer-associated tropomyosin 3.1 (Tpm3.1) provides the ability to examine the metabolic function of specific actin filament populations. We have determined the ability of these anti-Tpm (ATM) compounds to regulate glucose metabolism in mice. Acute treatment (1 h) of wild-type (WT) mice with the compounds (TR100 and ATM1001) led to a decrease in glucose clearance due mainly to suppression of glucose-stimulated insulin secretion (GSIS) from the pancreatic islets. The impact of the drugs on GSIS was significantly less in Tpm3.1 knock out (KO) mice indicating that the drug action is on-target. Experiments in MIN6 β-cells indicated that the inhibition of GSIS by the drugs was due to disruption to the cortical actin cytoskeleton. The impact of the drugs on insulin-stimulated glucose uptake (ISGU) was also examined in skeletal muscle ex vivo. In the absence of drug, ISGU was decreased in KO compared to WT muscle, confirming a role of Tpm3.1 in glucose uptake. Both compounds suppressed ISGU in WT muscle, but in the KO muscle there was little impact of the drugs. Collectively, this data indicates that the ATM drugs affect glucose metabolism in vivo by inhibiting Tpm3.1's function with few off-target effects.
Collapse
Affiliation(s)
- Anthony J Kee
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Jayshan Chagan
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Jeng Yie Chan
- Garvan Institute of Medical Research, St Vincent's Hospital, UNSW Sydney, Sydney, NSW, Australia
| | - Nicole S Bryce
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Christine A Lucas
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Jun Zeng
- MedChemSoft Solutions, Level 3 Brandon Park Drive, Wheelers Hill, 3150, VIC, Australia
| | - Jeff Hook
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Herbert Treutlein
- Sanoosa Pty. Ltd., 35 Collins Street, Melbourne, 3000, VIC, Australia
| | - D Ross Laybutt
- Garvan Institute of Medical Research, St Vincent's Hospital, UNSW Sydney, Sydney, NSW, Australia
| | - Justine R Stehn
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
- Novogen Pty Ltd, 502/20 George St, Hornsby, NSW, 2077, Australia
| | - Peter W Gunning
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Edna C Hardeman
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
29
|
Gonzalez JM, Ko MK, Hong YK, Weigert R, Tan JCH. Deep tissue analysis of distal aqueous drainage structures and contractile features. Sci Rep 2017; 7:17071. [PMID: 29213129 PMCID: PMC5719038 DOI: 10.1038/s41598-017-16897-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/15/2017] [Indexed: 01/04/2023] Open
Abstract
Outflow resistance in the aqueous drainage tract distal to trabecular meshwork is potentially an important determinant of intraocular pressure and success of trabecular bypass glaucoma surgeries. It is unclear how distal resistance is modulated. We sought to establish: (a) multimodal 2-photon deep tissue imaging and 3-dimensional analysis of the distal aqueous drainage tract (DT) in transgenic mice in vivo and ex vivo; (b) criteria for distinguishing the DT from blood and lymphatic vessels; and (c) presence of a DT wall organization capable of contractility. DT lumen appeared as scleral collagen second harmonic generation signal voids that could be traced back to Schlemm's canal. DT endothelium was Prox1-positive, CD31-positive and LYVE-1-negative, bearing a different molecular signature from blood and true lymphatic vessels. DT walls showed prominent filamentous actin (F-actin) labeling reflecting cells in a contracted state. F-actin co-localized with mesenchymal smooth muscle epitopes of alpha-smooth muscle actin, caldesmon and calponin, which localized adjacent and external to the endothelium. Our findings support a DT wall organization resembling that of blood vessels. This reflects a capacity to contract and support dynamic alteration of DT caliber and resistance analogous to the role of blood vessel tone in regulating blood flow.
Collapse
Affiliation(s)
- Jose M Gonzalez
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Minhee K Ko
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Young-Kwon Hong
- Department of Surgery, Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Robert Weigert
- Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - James C H Tan
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Ebrahim S, Milberg O, Weigert R. Isoform-specific roles of NMII drive membrane remodeling in vivo. Cell Cycle 2017; 16:1851-1852. [PMID: 28937881 DOI: 10.1080/15384101.2017.1372545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Seham Ebrahim
- a Laboratory of Cellular and Molecular Biology, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Oleg Milberg
- a Laboratory of Cellular and Molecular Biology, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Roberto Weigert
- a Laboratory of Cellular and Molecular Biology, Center for Cancer Research , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
31
|
Milberg O, Shitara A, Ebrahim S, Masedunskas A, Tora M, Tran DT, Chen Y, Conti MA, Adelstein RS, Ten Hagen KG, Weigert R. Concerted actions of distinct nonmuscle myosin II isoforms drive intracellular membrane remodeling in live animals. J Cell Biol 2017; 216:1925-1936. [PMID: 28600434 PMCID: PMC5496622 DOI: 10.1083/jcb.201612126] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/02/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022] Open
Abstract
Membrane remodeling plays a fundamental role during a variety of biological events. However, the dynamics and the molecular mechanisms regulating this process within cells in mammalian tissues in situ remain largely unknown. In this study, we use intravital subcellular microscopy in live mice to study the role of the actomyosin cytoskeleton in driving the remodeling of membranes of large secretory granules, which are integrated into the plasma membrane during regulated exocytosis. We show that two isoforms of nonmuscle myosin II, NMIIA and NMIIB, control distinct steps of the integration process. Furthermore, we find that F-actin is not essential for the recruitment of NMII to the secretory granules but plays a key role in the assembly and activation of NMII into contractile filaments. Our data support a dual role for the actomyosin cytoskeleton in providing the mechanical forces required to remodel the lipid bilayer and serving as a scaffold to recruit key regulatory molecules.
Collapse
Affiliation(s)
- Oleg Milberg
- Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| | - Akiko Shitara
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.,Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| | - Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Andrius Masedunskas
- Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD.,School of Medical Sciences, University of New South Wales, Sidney, Australia
| | - Muhibullah Tora
- Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| | - Duy T Tran
- Developmental Glycobiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Yun Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mary Anne Conti
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Kelly G Ten Hagen
- Developmental Glycobiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD .,Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| |
Collapse
|
32
|
Stoeckelhuber M, Loeffelbein DJ, Olzowy B, Schmitz C, Koerdt S, Kesting MR. Labial Salivary Glands in Infants: Histochemical Analysis of Cytoskeletal and Antimicrobial Proteins. J Histochem Cytochem 2017; 64:502-10. [PMID: 27439958 DOI: 10.1369/0022155416656940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
Human labial glands secrete mucous and serous substances for maintaining oral health. The normal microbial flora of the oral cavity is regulated by the acquired and innate immune systems. The localization and distribution of proteins of the innate immune system were investigated in serous acinar cells and the ductal system by the method of immunohistochemistry. Numerous antimicrobial proteins could be detected in the labial glands: β-defensin-1, -2, -3; lysozyme; lactoferrin; and cathelicidin. Cytoskeletal components such as actin, myosin II, cytokeratins 7 and 19, α- and β-tubulin were predominantly observed in apical cell regions and may be involved in secretory activities.
Collapse
Affiliation(s)
- Mechthild Stoeckelhuber
- Department of Oral and Maxillofacial Surgery, Technical University of Munich, Munich, Germany (MS, DJL, SK, MRK)
| | - Denys J Loeffelbein
- Department of Oral and Maxillofacial Surgery, Technical University of Munich, Munich, Germany (MS, DJL, SK, MRK)
| | - Bernhard Olzowy
- Department of Otorhinolaryngology, Ludwig Maximilians University of Munich, Munich, Germany (BO)
| | - Christoph Schmitz
- Department of Neuroanatomy, Ludwig Maximilians University of Munich, Munich, Germany (CS)
| | - Steffen Koerdt
- Department of Oral and Maxillofacial Surgery, Technical University of Munich, Munich, Germany (MS, DJL, SK, MRK)
| | - Marco R Kesting
- Department of Oral and Maxillofacial Surgery, Technical University of Munich, Munich, Germany (MS, DJL, SK, MRK)
| |
Collapse
|
33
|
Abstract
Exocytosis is an important cellular process controlled by metabolic signaling. It involves vesicle fusion to the plasma membrane, followed by the opening of a fusion pore, and the subsequent release of the vesicular lumen content into the extracellular space. While most modeling efforts focus on the events leading to membrane fusion, how the vesicular membrane remodels after fusing to plasma membrane remains unclear. This latter event dictates the nature and the efficiency of exocytotic vesicular secretions, and is thus critical for exocytotic function. We provide a generic membrane mechanical model to systematically study the fate of post-fusion vesicles. We show that while membrane stiffness favors full-collapse vesicle fusion into the plasma membrane, the intravesicular pressure swells the vesicle and causes the fusion pore to shrink. Dimensions of the vesicle and its associated fusion pore further modulate this mechanical antagonism. We systematically define the mechanical conditions that account for the full spectrum of the observed vesicular secretion modes. Our model therefore can serve as a unified theoretical framework that sheds light on the elaborate control mechanism of exocytosis.
Collapse
Affiliation(s)
- Thomas Stephens
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States of America. Equal contribution
| | | | | |
Collapse
|
34
|
Abstract
Exocytosis is a fundamental cellular process whereby secreted molecules are packaged into vesicles that move along cytoskeletal filaments and fuse with the plasma membrane. To function optimally, cells are strongly dependent on precisely controlled delivery of exocytotic cargo. In mammalian cells, microtubules serve as major tracks for vesicle transport by motor proteins, and thus microtubule organization is important for targeted delivery of secretory carriers. Over the years, multiple microtubule-associated and cortical proteins have been discovered that facilitate the interaction between the microtubule plus ends and the cell cortex. In this review, we focus on mammalian protein complexes that have been shown to participate in both cortical microtubule capture and exocytosis, thereby regulating the spatial organization of secretion. These complexes include microtubule plus-end tracking proteins, scaffolding factors, actin-binding proteins, and components of vesicle docking machinery, which together allow efficient coordination of cargo transport and release.
Collapse
Affiliation(s)
- Ivar Noordstra
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| |
Collapse
|
35
|
Abstract
Real-time imaging of regulated exocytosis in secreting organs can provide unprecedented temporal and spatial detail. Here, we highlight recent advances in 3D time-lapse imaging in Drosophila salivary glands at single-granule resolution. Using fluorescently labeled proteins expressed in the fly, it is now possible to image the dynamics of vesicle biogenesis and the cytoskeletal factors involved in secretion. 3D imaging over time allows one to visualize and define the temporal sequence of events, including clearance of cortical actin, fusion pore formation, mixing of the vesicular and plasma membranes and recruitment of components of the cytoskeleton. We will also discuss the genetic tools available in the fly that allow one to interrogate the essential factors involved in secretory vesicle formation, cargo secretion and the ultimate integration of the vesicular and plasma membranes. We argue that the combination of high-resolution real-time imaging and powerful genetics provides a platform to investigate the role of any factor in regulated secretion.
Collapse
Affiliation(s)
- Duy T Tran
- Section on Biological Chemistry, NIDCR, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Kelly G Ten Hagen
- Developmental Glycobiology Section, NIDCR, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Gluck C, Min S, Oyelakin A, Smalley K, Sinha S, Romano RA. RNA-seq based transcriptomic map reveals new insights into mouse salivary gland development and maturation. BMC Genomics 2016; 17:923. [PMID: 27852218 PMCID: PMC5112738 DOI: 10.1186/s12864-016-3228-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/29/2016] [Indexed: 11/16/2022] Open
Abstract
Background Mouse models have served a valuable role in deciphering various facets of Salivary Gland (SG) biology, from normal developmental programs to diseased states. To facilitate such studies, gene expression profiling maps have been generated for various stages of SG organogenesis. However these prior studies fall short of capturing the transcriptional complexity due to the limited scope of gene-centric microarray-based technology. Compared to microarray, RNA-sequencing (RNA-seq) offers unbiased detection of novel transcripts, broader dynamic range and high specificity and sensitivity for detection of genes, transcripts, and differential gene expression. Although RNA-seq data, particularly under the auspices of the ENCODE project, have covered a large number of biological specimens, studies on the SG have been lacking. Results To better appreciate the wide spectrum of gene expression profiles, we isolated RNA from mouse submandibular salivary glands at different embryonic and adult stages. In parallel, we processed RNA-seq data for 24 organs and tissues obtained from the mouse ENCODE consortium and calculated the average gene expression values. To identify molecular players and pathways likely to be relevant for SG biology, we performed functional gene enrichment analysis, network construction and hierarchal clustering of the RNA-seq datasets obtained from different stages of SG development and maturation, and other mouse organs and tissues. Our bioinformatics-based data analysis not only reaffirmed known modulators of SG morphogenesis but revealed novel transcription factors and signaling pathways unique to mouse SG biology and function. Finally we demonstrated that the unique SG gene signature obtained from our mouse studies is also well conserved and can demarcate features of the human SG transcriptome that is different from other tissues. Conclusions Our RNA-seq based Atlas has revealed a high-resolution cartographic view of the dynamic transcriptomic landscape of the mouse SG at various stages. These RNA-seq datasets will complement pre-existing microarray based datasets, including the Salivary Gland Molecular Anatomy Project by offering a broader systems-biology based perspective rather than the classical gene-centric view. Ultimately such resources will be valuable in providing a useful toolkit to better understand how the diverse cell population of the SG are organized and controlled during development and differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3228-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christian Gluck
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Sangwon Min
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Akinsola Oyelakin
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Kirsten Smalley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA.
| |
Collapse
|
37
|
Wang Z. Imaging Nanotherapeutics in Inflamed Vasculature by Intravital Microscopy. Am J Cancer Res 2016; 6:2431-2438. [PMID: 27877245 PMCID: PMC5118605 DOI: 10.7150/thno.16307] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/04/2016] [Indexed: 12/21/2022] Open
Abstract
Intravital microscopy (IVM) is the application of light microscopy to real time study biology of live animal tissues in intact and physiological conditions with the high spatial and temporal resolution. Advances in imaging systems, genetic animal models and imaging probes, IVM has offered quantitative and dynamic insight into cell biology, immunology, neurobiology and cancer. In this review, we will focus on the targeting of nanotherapeutics to inflamed vasculature. We will introduce the basic concept and principle of IVM and demonstrate that IVM is a powerful tool used to quantitatively determine the molecular mechanisms of interactions between nanotherapeutics and neutrophils or endothelium in living mice. In the future, it is needed to develop new imaging systems and novel imaging contrast agents to better understand molecular mechanisms of tissue processing of nanotherapeutics in vivo.
Collapse
|
38
|
Porat‐Shliom N, Tietgens AJ, Van Itallie CM, Vitale‐Cross L, Jarnik M, Harding OJ, Anderson JM, Gutkind JS, Weigert R, Arias IM. Liver kinase B1 regulates hepatocellular tight junction distribution and function in vivo. Hepatology 2016; 64:1317-29. [PMID: 27396550 PMCID: PMC5033699 DOI: 10.1002/hep.28724] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED Liver kinase B1 (LKB1) and its downstream effector AMP-activated protein kinase (AMPK) play critical roles in polarity establishment by regulating membrane trafficking and energy metabolism. In collagen sandwich-cultured hepatocytes, loss of LKB1 or AMPK impaired apical ABCB11 (Bsep) trafficking and bile canalicular formation. In the present study, we used liver-specific (albumin-Cre) LKB1 knockout mice (LKB1(-/-) ) to investigate the role of LKB1 in the maintenance of functional tight junction (TJ) in vivo. Transmission electron microscopy examination revealed that hepatocyte apical membrane with microvilli substantially extended into the basolateral domain of LKB1(-/-) livers. Immunofluorescence studies revealed that loss of LKB1 led to longer and wider canalicular structures correlating with mislocalization of the junctional protein, cingulin. To test junctional function, we used intravital microscopy to quantify the transport kinetics of 6-carboxyfluorescein diacetate (6-CFDA), which is processed in hepatocytes into its fluorescent derivative 6-carboxyfluorescein (6-CF) and secreted into the canaliculi. In LKB1(-/-) mice, 6-CF remained largely in hepatocytes, canalicular secretion was delayed, and 6-CF appeared in the blood. To test whether 6-CF was transported through permeable TJ, we intravenously injected low molecular weight (3 kDa) dextran in combination with 6-CFDA. In wild-type mice, 3 kDa dextran remained in the vasculature, whereas it rapidly appeared in the abnormal bile canaliculi in LKB1(-/-) mice, confirming that junctional disruption resulted in paracellular exchange between the blood stream and the bile canaliculus. CONCLUSION LKB1 plays a critical role in regulating the maintenance of TJ and paracellular permeability, which may explain how various drugs, chemicals, and metabolic states that inhibit the LKB1/AMPK pathway result in cholestasis. (Hepatology 2016;64:1317-1329).
Collapse
Affiliation(s)
- Natalie Porat‐Shliom
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMD,Laboratory of Cellular and Molecular Biology, National Cancer InstituteNational Institutes of HealthBethesdaMD
| | - Amber J. Tietgens
- Laboratory of Tight Junction Structure and FunctionNational Heart, Lung, and Blood InstituteBethesdaMD
| | - Christina M. Van Itallie
- Laboratory of Tight Junction Structure and FunctionNational Heart, Lung, and Blood InstituteBethesdaMD
| | - Lynn Vitale‐Cross
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMD
| | - Michal Jarnik
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMD
| | - Olivia J. Harding
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMD
| | - James M. Anderson
- Laboratory of Tight Junction Structure and FunctionNational Heart, Lung, and Blood InstituteBethesdaMD
| | - J. Silvio Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMD,Present address: University of California, San Diego, Moores Cancer CenterLa JollaCA92093
| | - Roberto Weigert
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMD,Laboratory of Cellular and Molecular Biology, National Cancer InstituteNational Institutes of HealthBethesdaMD
| | - Irwin M. Arias
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMD
| |
Collapse
|
39
|
Meunier FA, Gutiérrez LM. Captivating New Roles of F-Actin Cortex in Exocytosis and Bulk Endocytosis in Neurosecretory Cells. Trends Neurosci 2016; 39:605-613. [DOI: 10.1016/j.tins.2016.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/01/2016] [Accepted: 07/06/2016] [Indexed: 12/01/2022]
|
40
|
Wen PJ, Grenklo S, Arpino G, Tan X, Liao HS, Heureaux J, Peng SY, Chiang HC, Hamid E, Zhao WD, Shin W, Näreoja T, Evergren E, Jin Y, Karlsson R, Ebert SN, Jin A, Liu AP, Shupliakov O, Wu LG. Actin dynamics provides membrane tension to merge fusing vesicles into the plasma membrane. Nat Commun 2016; 7:12604. [PMID: 27576662 PMCID: PMC5013665 DOI: 10.1038/ncomms12604] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 07/13/2016] [Indexed: 01/22/2023] Open
Abstract
Vesicle fusion is executed via formation of an Ω-shaped structure (Ω-profile), followed by closure (kiss-and-run) or merging of the Ω-profile into the plasma membrane (full fusion). Although Ω-profile closure limits release but recycles vesicles economically, Ω-profile merging facilitates release but couples to classical endocytosis for recycling. Despite its crucial role in determining exocytosis/endocytosis modes, how Ω-profile merging is mediated is poorly understood in endocrine cells and neurons containing small ∼30–300 nm vesicles. Here, using confocal and super-resolution STED imaging, force measurements, pharmacology and gene knockout, we show that dynamic assembly of filamentous actin, involving ATP hydrolysis, N-WASP and formin, mediates Ω-profile merging by providing sufficient plasma membrane tension to shrink the Ω-profile in neuroendocrine chromaffin cells containing ∼300 nm vesicles. Actin-directed compounds also induce Ω-profile accumulation at lamprey synaptic active zones, suggesting that actin may mediate Ω-profile merging at synapses. These results uncover molecular and biophysical mechanisms underlying Ω-profile merging. As vesicles fuse to the plasma membrane, they form intermediate Ω-shaped structures followed by either closure of the pore or full merging with the plasma membrane. Here Wen et al. show that dynamic actin assembly provides membrane tension to promote Ω merging in neuroendocrine cells and synapses.
Collapse
Affiliation(s)
- Peter J Wen
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Staffan Grenklo
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden.,Department of Cell Biology, WGI, Stockholm University, 106 91 Stockholm, Sweden
| | - Gianvito Arpino
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA.,Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Xinyu Tan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Hsien-Shun Liao
- National Institute of Biomedical Imaging and Bioengineering (NIBIB), Bethesda, Maryland 20892, USA
| | - Johanna Heureaux
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Shi-Yong Peng
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Hsueh-Cheng Chiang
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Edaeni Hamid
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Wei-Dong Zhao
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Wonchul Shin
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Tuomas Näreoja
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Emma Evergren
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Yinghui Jin
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| | - Roger Karlsson
- Department of Cell Biology, WGI, Stockholm University, 106 91 Stockholm, Sweden
| | - Steven N Ebert
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, Florida 32827, USA
| | - Albert Jin
- National Institute of Biomedical Imaging and Bioengineering (NIBIB), Bethesda, Maryland 20892, USA
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Oleg Shupliakov
- Center of Excellence in Developmental Biology, Department of Neuroscience, Karolinska Institutet, S-171 77 Stockholm, Sweden.,Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Building 35, Room 2B-1012, Bethesda, Maryland 20892, USA
| |
Collapse
|
41
|
Gonzalez JM, Ko MK, Masedunskas A, Hong YK, Weigert R, Tan JCH. Toward in vivo two-photon analysis of mouse aqueous outflow structure and function. Exp Eye Res 2016; 158:161-170. [PMID: 27179411 DOI: 10.1016/j.exer.2016.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 12/29/2022]
Abstract
The promise of revolutionary insights into intraocular pressure (IOP) and aqueous humor outflow homeostasis, IOP pathogenesis, and novel therapy offered by engineered mouse models has been hindered by a lack of appropriate tools for studying the aqueous drainage tissues in their original 3-dimensional (3D) environment. Advances in 2-photon excitation fluorescence imaging (TPEF) combined with availability of modalities such as transgenic reporter mice and intravital dyes have placed us on the cusp of unlocking the potential of the mouse model for unearthing insights into aqueous drainage structure and function. Multimodality 2-photon imaging permits high-resolution visualization not only of tissue structural organization but also cells and cellular function. It is possible to dig deeper into understanding the cellular basis of aqueous outflow regulation as the technique integrates analysis of tissue structure, cell biology and physiology in a way that could also lead to fresh insights into human glaucoma. We outline recent novel applications of two-photon imaging to analyze the mouse conventional drainage system in vivo or in whole tissues: (1) collagen second harmonic generation (SHG) identifies the locations of episcleral vessels, intrascleral plexuses, collector channels, and Schlemm's canal in the distal aqueous drainage tract; (2) the prospero homeobox protein 1-green fluorescent protein (GFP) reporter helps locate the inner wall of Schlemm's canal; (3) Calcein AM, siGLO™, the fluorescent reporters m-Tomato and GFP, and coherent anti-Stokes scattering (CARS), are adjuncts to TPEF to identify live cells by their membrane or cytosolic locations; (4) autofluorescence and sulforhodamine-B to identify elastic fibers in the living eye. These tools greatly expand our options for analyzing physiological and pathological processes in the aqueous drainage tissues of live mice as a model of the analogous human system.
Collapse
Affiliation(s)
- Jose M Gonzalez
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Minhee K Ko
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Andrius Masedunskas
- Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Young-Kwon Hong
- Department of Surgery, Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roberto Weigert
- Intracellular Membrane Trafficking Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - James C H Tan
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
42
|
Tanaka S, Takakuwa Y. A method for detailed analysis of the structure of mast cell secretory granules by negative contrast imaging. Sci Rep 2016; 6:23369. [PMID: 26997316 PMCID: PMC4800307 DOI: 10.1038/srep23369] [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: 08/25/2015] [Accepted: 03/04/2016] [Indexed: 12/15/2022] Open
Abstract
Secretory granules (SGs) in mast cells contain various molecules that elicit allergy symptoms and are generally considered therapeutic targets. However, the biogenesis, maintenance, regulation, and recycling of these granules remain controversial, mainly due to the lack of suitable live-cell imaging methods. In this study, we applied negative contrast imaging with soluble green fluorescent protein (GFP) expressed in the cytoplasm as a method to validate structural information of mast cell SGs. We evaluated the accuracy of the method in detail, and we demonstrated that it can be used for quantitative analysis. Using this technique, secretory granules, the nucleus, mitochondria, and the cell body were visualized in individual RBL-2H3 mast cells without any influence. When combined with conventional multicolor fluorescence imaging, visualization of SG-associated proteins and SG-SG fusion was achieved. Moreover, 3D images were constructed based on this method, and detailed information on the number, size, and shape of individual SGs was obtained. We found that cell volume was correlated with SG number. In summary, the technique provides valuable and unique data, and will therefore advance SG research.
Collapse
Affiliation(s)
- Shotaro Tanaka
- Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Kawada 8-1, Shinjuku, Tokyo 162-8666, Japan
| | - Yuichi Takakuwa
- Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Kawada 8-1, Shinjuku, Tokyo 162-8666, Japan
| |
Collapse
|
43
|
Baker OJ. Current trends in salivary gland tight junctions. Tissue Barriers 2016; 4:e1162348. [PMID: 27583188 DOI: 10.1080/21688370.2016.1162348] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/28/2016] [Accepted: 03/01/2016] [Indexed: 12/21/2022] Open
Abstract
Tight junctions form a continuous intercellular barrier between epithelial cells that is required to separate tissue spaces and regulate selective movement of solutes across the epithelium. They are composed of strands containing integral membrane proteins (e.g., claudins, occludin and tricellulin, junctional adhesion molecules and the coxsackie adenovirus receptor). These proteins are anchored to the cytoskeleton via scaffolding proteins such as ZO-1 and ZO-2. In salivary glands, tight junctions are involved in polarized saliva secretion and barrier maintenance between the extracellular environment and the glandular lumen. This review seeks to provide an overview of what is currently known, as well as the major questions and future research directions, regarding tight junction expression, organization and function within salivary glands.
Collapse
Affiliation(s)
- Olga J Baker
- School of Dentistry, University of Utah , Salt Lake City, UT, USA
| |
Collapse
|
44
|
Kittelberger N, Breunig M, Martin R, Knölker HJ, Miklavc P. The role of myosin 1c and myosin 1b in surfactant exocytosis. J Cell Sci 2016; 129:1685-96. [PMID: 26940917 PMCID: PMC4852769 DOI: 10.1242/jcs.181313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/25/2016] [Indexed: 12/19/2022] Open
Abstract
Actin and actin-associated proteins have a pivotal effect on regulated exocytosis in secretory cells and influence pre-fusion as well as post-fusion stages of exocytosis. Actin polymerization on secretory granules during the post-fusion phase (formation of an actin coat) is especially important in cells with large secretory vesicles or poorly soluble secretions. Alveolar type II (ATII) cells secrete hydrophobic lipo-protein surfactant, which does not easily diffuse from fused vesicles. Previous work showed that compression of actin coat is necessary for surfactant extrusion. Here, we investigate the role of class 1 myosins as possible linkers between actin and membranes during exocytosis. Live-cell microscopy showed translocation of fluorescently labeled myosin 1b and myosin 1c to the secretory vesicle membrane after fusion. Myosin 1c translocation was dependent on its pleckstrin homology domain. Expression of myosin 1b and myosin 1c constructs influenced vesicle compression rate, whereas only the inhibition of myosin 1c reduced exocytosis. These findings suggest that class 1 myosins participate in several stages of ATII cell exocytosis and link actin coats to the secretory vesicle membrane to influence vesicle compression.
Collapse
Affiliation(s)
- Nadine Kittelberger
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| | - Markus Breunig
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| | - René Martin
- Department of Chemistry, Technische Universität Dresden, Bergstr. 66, Dresden 01069, Germany
| | - Hans-Joachim Knölker
- Department of Chemistry, Technische Universität Dresden, Bergstr. 66, Dresden 01069, Germany
| | - Pika Miklavc
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| |
Collapse
|
45
|
Arp2/3-mediated F-actin formation controls regulated exocytosis in vivo. Nat Commun 2015; 6:10098. [PMID: 26639106 PMCID: PMC4686765 DOI: 10.1038/ncomms10098] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/02/2015] [Indexed: 02/07/2023] Open
Abstract
The actin cytoskeleton plays crucial roles in many cellular processes, including regulated secretion. However, the mechanisms controlling F-actin dynamics in this process are largely unknown. Through 3D time-lapse imaging in a secreting organ, we show that F-actin is actively disassembled along the apical plasma membrane at the site of secretory vesicle fusion and re-assembled directionally on vesicle membranes. Moreover, we show that fusion pore formation and PIP2 redistribution precedes actin and myosin recruitment to secretory vesicle membranes. Finally, we show essential roles for the branched actin nucleators Arp2/3- and WASp in the process of secretory cargo expulsion and integration of vesicular membranes with the apical plasma membrane. Our results highlight previously unknown roles for branched actin in exocytosis and provide a genetically tractable system to image the temporal and spatial dynamics of polarized secretion in vivo. The cytoskeleton plays a crucial role in secretion. Here Tran et al. demonstrate that cortical actin is rearranged at the site of vesicle fusion and recruited to fused secretory granules in Drosophila salivary glands, and show that branched actin nucleators are required for cargo expulsion.
Collapse
|
46
|
Orchestrated content release from Drosophila glue-protein vesicles by a contractile actomyosin network. Nat Cell Biol 2015; 18:181-90. [DOI: 10.1038/ncb3288] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/11/2015] [Indexed: 12/22/2022]
|
47
|
Arous C, Halban PA. The skeleton in the closet: actin cytoskeletal remodeling in β-cell function. Am J Physiol Endocrinol Metab 2015; 309:E611-20. [PMID: 26286869 DOI: 10.1152/ajpendo.00268.2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/11/2015] [Indexed: 01/13/2023]
Abstract
Over the last few decades, biomedical research has considered not only the function of single cells but also the importance of the physical environment within a whole tissue, including cell-cell and cell-extracellular matrix interactions. Cytoskeleton organization and focal adhesions are crucial sensors for cells that enable them to rapidly communicate with the physical extracellular environment in response to extracellular stimuli, ensuring proper function and adaptation. The involvement of the microtubular-microfilamentous cytoskeleton in secretion mechanisms was proposed almost 50 years ago, since when the evolution of ever more sensitive and sophisticated methods in microscopy and in cell and molecular biology have led us to become aware of the importance of cytoskeleton remodeling for cell shape regulation and its crucial link with signaling pathways leading to β-cell function. Emerging evidence suggests that dysfunction of cytoskeletal components or extracellular matrix modification influences a number of disorders through potential actin cytoskeleton disruption that could be involved in the initiation of multiple cellular functions. Perturbation of β-cell actin cytoskeleton remodeling could arise secondarily to islet inflammation and fibrosis, possibly accounting in part for impaired β-cell function in type 2 diabetes. This review focuses on the role of actin remodeling in insulin secretion mechanisms and its close relationship with focal adhesions and myosin II.
Collapse
Affiliation(s)
- Caroline Arous
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| | - Philippe A Halban
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
48
|
Shitara A, Weigert R. Imaging membrane remodeling during regulated exocytosis in live mice. Exp Cell Res 2015; 337:219-25. [PMID: 26160452 DOI: 10.1016/j.yexcr.2015.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 06/28/2015] [Indexed: 10/23/2022]
Abstract
In this mini-review we focus on the use of time-lapse light microscopy to study membrane remodeling during protein secretion in live animals. In particular, we highlight how subcellular intravital microscopy has enabled imaging the dynamics of both individual secretory vesicles and the plasma membrane, during different steps in the exocytic process. This powerful approach has provided us with the unique opportunity to unravel the role of the actin cytoskeleton in regulating this process under physiological conditions, and to overcome the shortcomings of more reductionist model systems.
Collapse
Affiliation(s)
- Akiko Shitara
- Intracellular Membrane Trafficking Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Dr. 303A, Bethesda, MD 20892-4340, United States
| | - Roberto Weigert
- Intracellular Membrane Trafficking Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Dr. 303A, Bethesda, MD 20892-4340, United States.
| |
Collapse
|
49
|
Zaidel-Bar R, Zhenhuan G, Luxenburg C. The contractome – a systems view of actomyosin contractility in non-muscle cells. J Cell Sci 2015; 128:2209-17. [DOI: 10.1242/jcs.170068] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/27/2015] [Indexed: 12/21/2022] Open
Abstract
ABSTRACT
Actomyosin contractility is a highly regulated process that affects many fundamental biological processes in each and every cell in our body. In this Cell Science at a Glance article and the accompanying poster, we mined the literature and databases to map the contractome of non-muscle cells. Actomyosin contractility is involved in at least 49 distinct cellular functions that range from providing cell architecture to signal transduction and nuclear activity. Containing over 100 scaffolding and regulatory proteins, the contractome forms a highly complex network with more than 230 direct interactions between its components, 86 of them involving phosphorylation. Mapping these interactions, we identify the key regulatory pathways involved in the assembly of actomyosin structures and in activating myosin to produce contractile forces within non-muscle cells at the exact time and place necessary for cellular function.
Collapse
Affiliation(s)
- Ronen Zaidel-Bar
- Mechanobiology Institute, National University of Singapore, T-lab building #05-01, 5A Engineering Drive 1, 117411, Singapore
| | - Guo Zhenhuan
- Mechanobiology Institute, National University of Singapore, T-lab building #05-01, 5A Engineering Drive 1, 117411, Singapore
| | - Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, P.O. Box 39040, Tel Aviv 69978, Israel
| |
Collapse
|
50
|
Nezu A, Morita T, Tanimura A. In vitro and in vivo imaging of intracellular Ca2+ responses in salivary gland cells. J Oral Biosci 2015. [DOI: 10.1016/j.job.2015.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|