1
|
Liboy-Lugo JM, Espinoza CA, Sheu-Gruttadauria J, Park JE, Xu A, Jowhar Z, Gao AL, Carmona-Negrón JA, Wittmann T, Jura N, Floor SN. G3BP isoforms differentially affect stress granule assembly and gene expression during cellular stress. Mol Biol Cell 2024; 35:ar140. [PMID: 39356796 DOI: 10.1091/mbc.e24-02-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
Stress granules (SGs) are macromolecular assemblies that form under cellular stress. Formation of these membraneless organelles is driven by the condensation of RNA and RNA-binding proteins such as G3BPs. G3BPs form SGs following stress-induced translational arrest. Three G3BP paralogues (G3BP1, G3BP2A, and G3BP2B) have been identified in vertebrates. However, the contribution of different G3BP paralogues to SG formation and gene expression changes is incompletely understood. Here, we probed the functions of G3BPs by identifying important residues for SG assembly at their N-terminal domain such as V11. This conserved amino acid is required for formation of the G3BP-Caprin-1 complex, hence promoting SG assembly. Total RNA sequencing and ribosome profiling revealed that a G3BPV11A mutant leads to changes in mRNA levels and ribosome engagement during the integrated stress response (ISR). Moreover, we found that G3BP2B preferentially forms SGs and promotes changes in mRNA expression under endoplasmic reticulum (ER) stress. Furthermore, our work is a resource for researchers to study gene expression changes under cellular stress. Together, this work suggests that perturbing protein-protein interactions mediated by G3BPs affect SG assembly and gene expression during the ISR, and such functions are differentially regulated by G3BP paralogues under ER stress.
Collapse
Affiliation(s)
- José M Liboy-Lugo
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158
| | - Carla A Espinoza
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Jessica Sheu-Gruttadauria
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Jesslyn E Park
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Albert Xu
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Ziad Jowhar
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143
| | - Angela L Gao
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158
| | - José A Carmona-Negrón
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158
- Department of Chemistry, University of Puerto Rico, Mayagüez, PR 00680
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158
| | - Stephen N Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
2
|
Ju RJ, Falconer AD, Schmidt CJ, Enriquez Martinez MA, Dean KM, Fiolka RP, Sester DP, Nobis M, Timpson P, Lomakin AJ, Danuser G, White MD, Haass NK, Oelz DB, Stehbens SJ. Compression-dependent microtubule reinforcement enables cells to navigate confined environments. Nat Cell Biol 2024; 26:1520-1534. [PMID: 39160291 DOI: 10.1038/s41556-024-01476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/11/2024] [Indexed: 08/21/2024]
Abstract
Cells migrating through complex three-dimensional environments experience considerable physical challenges, including tensile stress and compression. To move, cells need to resist these forces while also squeezing the large nucleus through confined spaces. This requires highly coordinated cortical contractility. Microtubules can both resist compressive forces and sequester key actomyosin regulators to ensure appropriate activation of contractile forces. Yet, how these two roles are integrated to achieve nuclear transmigration in three dimensions is largely unknown. Here, we demonstrate that compression triggers reinforcement of a dedicated microtubule structure at the rear of the nucleus by the mechanoresponsive recruitment of cytoplasmic linker-associated proteins, which dynamically strengthens and repairs the lattice. These reinforced microtubules form the mechanostat: an adaptive feedback mechanism that allows the cell to both withstand compressive force and spatiotemporally organize contractility signalling pathways. The microtubule mechanostat facilitates nuclear positioning and coordinates force production to enable the cell to pass through constrictions. Disruption of the mechanostat imbalances cortical contractility, stalling migration and ultimately resulting in catastrophic cell rupture. Our findings reveal a role for microtubules as cellular sensors that detect and respond to compressive forces, enabling movement and ensuring survival in mechanically demanding environments.
Collapse
Affiliation(s)
- Robert J Ju
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Alistair D Falconer
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia
| | - Christanny J Schmidt
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Marco A Enriquez Martinez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Kevin M Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Reto P Fiolka
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David P Sester
- TRI Flow Cytometry Suite (TRI.fcs), Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Max Nobis
- Faculty of Medicine, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Paul Timpson
- Faculty of Medicine, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Alexis J Lomakin
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
- Institute of Medical Chemistry and Pathobiochemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melanie D White
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Nikolas K Haass
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Dietmar B Oelz
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia.
| | - Samantha J Stehbens
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
3
|
Folts L, Martinez AS, McKey J. Tissue clearing and imaging approaches for in toto analysis of the reproductive system†. Biol Reprod 2024; 110:1041-1054. [PMID: 38159104 PMCID: PMC11180619 DOI: 10.1093/biolre/ioad182] [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: 11/01/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024] Open
Abstract
New microscopy techniques in combination with tissue clearing protocols and emerging analytical approaches have presented researchers with the tools to understand dynamic biological processes in a three-dimensional context. This paves the road for the exploration of new research questions in reproductive biology, for which previous techniques have provided only approximate resolution. These new methodologies now allow for contextualized analysis of far-larger volumes than was previously possible. Tissue optical clearing and three-dimensional imaging techniques posit the bridging of molecular mechanisms, macroscopic morphogenic development, and maintenance of reproductive function into one cohesive and comprehensive understanding of the biology of the reproductive system. In this review, we present a survey of the various tissue clearing techniques and imaging systems, as they have been applied to the developing and adult reproductive system. We provide an overview of tools available for analysis of experimental data, giving particular attention to the emergence of artificial intelligence-assisted methods and their applicability to image analysis. We conclude with an evaluation of how novel image analysis approaches that have been applied to other organ systems could be incorporated into future experimental evaluation of reproductive biology.
Collapse
Affiliation(s)
- Lillian Folts
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO, USA
| | - Anthony S Martinez
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO, USA
| | - Jennifer McKey
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO, USA
| |
Collapse
|
4
|
Dema A, Charafeddine RA, van Haren J, Rahgozar S, Viola G, Jacobs KA, Kutys ML, Wittmann T. Doublecortin reinforces microtubules to promote growth cone advance in soft environments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582626. [PMID: 38464100 PMCID: PMC10925279 DOI: 10.1101/2024.02.28.582626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Doublecortin (DCX) is a microtubule-associated protein critical for brain development. Although most highly expressed in the developing central nervous system, the molecular function of DCX in neuron morphogenesis remains unknown and controversial. We demonstrate that DCX function is intimately linked to its microtubule-binding activity. By using human induced pluripotent stem cell (hiPSC)- derived cortical i 3 Neurons genome engineered to express mEmerald-tagged DCX from the endogenous locus, we find that DCX-MT interactions become highly polarized very early during neuron morphogenesis. DCX becomes enriched only on straight microtubules in advancing growth cones with approximately 120 DCX molecules bound per micrometer of growth cone microtubule. At a similar saturation, microtubule-bound DCX molecules begin to impede lysosome transport, and thus can potentially control growth cone organelle entry. In addition, by comparing control, DCX-mEmerald and knockout DCX -/Y i 3 Neurons, we find that DCX stabilizes microtubules in the growth cone peripheral domain by reducing the microtubule catastrophe frequency and the depolymerization rate. DCX -/Y i 3 Neuron morphogenesis was inhibited in soft microenvironments that mimic the viscoelasticity of brain tissue and DCX -/Y neurites failed to grow toward brain-derived neurotrophic factor (BDNF) gradients. Together with high resolution traction force microscopy data, we propose a model in which DCX-decorated, rigid growth cone microtubules provide intracellular mechanical resistance to actomyosin generated contractile forces in soft physiological environments in which weak and transient adhesion-mediated forces in the growth cone periphery may be insufficient for productive growth cone advance. These data provide a new mechanistic understanding of how DCX mutations cause lissencephaly-spectrum brain malformations by impacting growth cone dynamics during neuron morphogenesis in physiological environments.
Collapse
|
5
|
Liboy-Lugo JM, Espinoza CA, Sheu-Gruttadauria J, Park JE, Xu A, Jowhar Z, Gao AL, Carmona-Negrón JA, Wittmann T, Jura N, Floor SN. Protein-protein interactions with G3BPs drive stress granule condensation and gene expression changes under cellular stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579149. [PMID: 38370785 PMCID: PMC10871250 DOI: 10.1101/2024.02.06.579149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Stress granules (SGs) are macromolecular assemblies that form under cellular stress. Formation of these condensates is driven by the condensation of RNA and RNA-binding proteins such as G3BPs. G3BPs condense into SGs following stress-induced translational arrest. Three G3BP paralogs (G3BP1, G3BP2A, and G3BP2B) have been identified in vertebrates. However, the contribution of different G3BP paralogs to stress granule formation and stress-induced gene expression changes is incompletely understood. Here, we identified key residues for G3BP condensation such as V11. This conserved amino acid is required for formation of the G3BP-Caprin-1 complex, hence promoting SG assembly. Total RNA sequencing and ribosome profiling revealed that disruption of G3BP condensation corresponds to changes in mRNA levels and ribosome engagement during the integrated stress response (ISR). Moreover, we found that G3BP2B preferentially condenses and promotes changes in mRNA expression under endoplasmic reticulum (ER) stress. Together, this work suggests that stress granule assembly promotes changes in gene expression under cellular stress, which is differentially regulated by G3BP paralogs.
Collapse
Affiliation(s)
- José M. Liboy-Lugo
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, California, USA
| | - Carla A. Espinoza
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA
| | - Jessica Sheu-Gruttadauria
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA
| | - Jesslyn E. Park
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
| | - Albert Xu
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
| | - Ziad Jowhar
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, California, USA
| | - Angela L. Gao
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, California, USA
| | - José A. Carmona-Negrón
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Chemistry, University of Puerto Rico, Mayaguez, Puerto Rico, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, California, USA
| | - Stephen N. Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
6
|
Smart K, Kramer AH, Smart S, Hodgson L, Sharp DJ. Fidgetin-like 2 depletion enhances cell migration by regulating GEF-H1, RhoA, and FAK. Biophys J 2023; 122:3600-3610. [PMID: 36523161 PMCID: PMC10541466 DOI: 10.1016/j.bpj.2022.12.018] [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: 08/22/2022] [Revised: 10/31/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The microtubule (MT) cytoskeleton and its dynamics play an important role in cell migration. Depletion of the microtubule-severing enzyme Fidgetin-like 2 (FL2), a regulator of MT dynamics at the leading edge of migrating cells, leads to faster and more efficient cell migration. Here we examine how siRNA knockdown of FL2 increases cell motility. Förster resonance energy transfer biosensor studies shows that FL2 knockdown decreases activation of the p21 Rho GTPase, RhoA, and its activator GEF-H1. Immunofluorescence studies reveal that GEF-H1 is sequestered by the increased MT density resulting from FL2 depletion. Activation of the Rho GTPase, Rac1, however, does not change after FL2 knockdown. Furthermore, FL2 depletion leads to an increase in focal adhesion kinase activation at the leading edge, as shown by immunofluorescence studies, but no change in actin dynamics, as shown by fluorescence recovery after photobleaching. We believe these results expand our understanding of the role of MT dynamics in cell migration and offer new insights into RhoA and Rac1 regulation.
Collapse
Affiliation(s)
- Karishma Smart
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Adam H Kramer
- Microcures, Inc., Research and Development, Bronx, New York
| | | | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York.
| | - David J Sharp
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York; Microcures, Inc., Research and Development, Bronx, New York.
| |
Collapse
|
7
|
Parker M, Annamdevula NS, Pleshinger D, Ijaz Z, Jalkh J, Penn R, Deshpande D, Rich TC, Leavesley SJ. Comparing Performance of Spectral Image Analysis Approaches for Detection of Cellular Signals in Time-Lapse Hyperspectral Imaging Fluorescence Excitation-Scanning Microscopy. Bioengineering (Basel) 2023; 10:642. [PMID: 37370573 PMCID: PMC10295298 DOI: 10.3390/bioengineering10060642] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Hyperspectral imaging (HSI) technology has been applied in a range of fields for target detection and mixture analysis. While HSI was originally developed for remote sensing applications, modern uses include agriculture, historical document authentication, and medicine. HSI has also shown great utility in fluorescence microscopy. However, traditional fluorescence microscopy HSI systems have suffered from limited signal strength due to the need to filter or disperse the emitted light across many spectral bands. We have previously demonstrated that sampling the fluorescence excitation spectrum may provide an alternative approach with improved signal strength. Here, we report on the use of excitation-scanning HSI for dynamic cell signaling studies-in this case, the study of the second messenger Ca2+. Time-lapse excitation-scanning HSI data of Ca2+ signals in human airway smooth muscle cells (HASMCs) were acquired and analyzed using four spectral analysis algorithms: linear unmixing (LU), spectral angle mapper (SAM), constrained energy minimization (CEM), and matched filter (MF), and the performances were compared. Results indicate that LU and MF provided similar linear responses to increasing Ca2+ and could both be effectively used for excitation-scanning HSI. A theoretical sensitivity framework was used to enable the filtering of analyzed images to reject pixels with signals below a minimum detectable limit. The results indicated that subtle kinetic features might be revealed through pixel filtering. Overall, the results suggest that excitation-scanning HSI can be employed for kinetic measurements of cell signals or other dynamic cellular events and that the selection of an appropriate analysis algorithm and pixel filtering may aid in the extraction of quantitative signal traces. These approaches may be especially helpful for cases where the signal of interest is masked by strong cellular autofluorescence or other competing signals.
Collapse
Affiliation(s)
- Marina Parker
- Department of Chemical and Biomolecular Engineering, University of South Alabama, 150 Student Services Dr., Mobile, AL 36688, USA
- Department of Systems Engineering, University of South Alabama, 150 Student Services Dr., Mobile, AL 36688, USA
| | - Naga S. Annamdevula
- Department of Pharmacology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA; (N.S.A.)
- Center for Lung Biology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA
| | - Donald Pleshinger
- Department of Pharmacology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA; (N.S.A.)
- Center for Lung Biology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA
| | - Zara Ijaz
- College of Medicine, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA
| | - Josephine Jalkh
- College of Medicine, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA
| | - Raymond Penn
- College of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Deepak Deshpande
- College of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Thomas C. Rich
- Department of Pharmacology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA; (N.S.A.)
- Center for Lung Biology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA
| | - Silas J. Leavesley
- Department of Chemical and Biomolecular Engineering, University of South Alabama, 150 Student Services Dr., Mobile, AL 36688, USA
- Department of Systems Engineering, University of South Alabama, 150 Student Services Dr., Mobile, AL 36688, USA
- Department of Pharmacology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA; (N.S.A.)
- Center for Lung Biology, University of South Alabama, 5851 USA Drive N., Mobile, AL 36688, USA
| |
Collapse
|
8
|
Dema A, Charafeddine R, Rahgozar S, van Haren J, Wittmann T. Growth cone advance requires EB1 as revealed by genomic replacement with a light-sensitive variant. eLife 2023; 12:84143. [PMID: 36715499 PMCID: PMC9917429 DOI: 10.7554/elife.84143] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A challenge in analyzing dynamic intracellular cell biological processes is the dearth of methodologies that are sufficiently fast and specific to perturb intracellular protein activities. We previously developed a light-sensitive variant of the microtubule plus end-tracking protein EB1 by inserting a blue light-controlled protein dimerization module between functional domains. Here, we describe an advanced method to replace endogenous EB1 with this light-sensitive variant in a single genome editing step, thereby enabling this approach in human induced pluripotent stem cells (hiPSCs) and hiPSC-derived neurons. We demonstrate that acute and local optogenetic EB1 inactivation in developing cortical neurons induces microtubule depolymerization in the growth cone periphery and subsequent neurite retraction. In addition, advancing growth cones are repelled from areas of blue light exposure. These phenotypes were independent of the neuronal EB1 homolog EB3, revealing a direct dynamic role of EB1-mediated microtubule plus end interactions in neuron morphogenesis and neurite guidance.
Collapse
Affiliation(s)
- Alessandro Dema
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | - Rabab Charafeddine
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | - Shima Rahgozar
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | | | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
9
|
Chauvistré H, Shannan B, Daignault-Mill SM, Ju RJ, Picard D, Egetemaier S, Váraljai R, Gibhardt CS, Sechi A, Kaschani F, Keminer O, Stehbens SJ, Liu Q, Yin X, Jeyakumar K, Vogel FCE, Krepler C, Rebecca VW, Kubat L, Lueong SS, Forster J, Horn S, Remke M, Ehrmann M, Paschen A, Becker JC, Helfrich I, Rauh D, Kaiser M, Gul S, Herlyn M, Bogeski I, Rodríguez-López JN, Haass NK, Schadendorf D, Roesch A. Persister state-directed transitioning and vulnerability in melanoma. Nat Commun 2022; 13:3055. [PMID: 35650266 PMCID: PMC9160289 DOI: 10.1038/s41467-022-30641-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/10/2022] [Indexed: 12/30/2022] Open
Abstract
Melanoma is a highly plastic tumor characterized by dynamic interconversion of different cell identities depending on the biological context. Melanoma cells with high expression of the H3K4 demethylase KDM5B (JARID1B) rest in a slow-cycling, yet reversible persister state. Over time, KDM5Bhigh cells can promote rapid tumor repopulation with equilibrated KDM5B expression heterogeneity. The cellular identity of KDM5Bhigh persister cells has not been studied so far, missing an important cell state-directed treatment opportunity in melanoma. Here, we have established a doxycycline-titratable system for genetic induction of permanent intratumor expression of KDM5B and screened for chemical agents that phenocopy this effect. Transcriptional profiling and cell functional assays confirmed that the dihydropyridine 2-phenoxyethyl 4-(2-fluorophenyl)-2,7,7-trimethyl-5-oxo-1,4,5,6,7,8-hexa-hydro-quinoline-3-carboxylate (termed Cpd1) supports high KDM5B expression and directs melanoma cells towards differentiation along the melanocytic lineage and to cell cycle-arrest. The high KDM5B state additionally prevents cell proliferation through negative regulation of cytokinetic abscission. Moreover, treatment with Cpd1 promoted the expression of the melanocyte-specific tyrosinase gene specifically sensitizing melanoma cells for the tyrosinase-processed antifolate prodrug 3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG). In summary, our study provides proof-of-concept for a dual hit strategy in melanoma, in which persister state-directed transitioning limits tumor plasticity and primes melanoma cells towards lineage-specific elimination.
Collapse
Affiliation(s)
- Heike Chauvistré
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Batool Shannan
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Sheena M Daignault-Mill
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Robert J Ju
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel Picard
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Stefanie Egetemaier
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Renáta Váraljai
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Antonio Sechi
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Farnusch Kaschani
- Department of Chemical Biology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
- Center for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, 22525, Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Schnackenburgallee 114, 22525, Hamburg, Germany
| | - Samantha J Stehbens
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Qin Liu
- The Wistar Institute, Philadelphia, PA, USA
| | | | - Kirujan Jeyakumar
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - Felix C E Vogel
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | | | | | - Linda Kubat
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), University Hospital of Essen, Universitätsstrasse 1, 45141, Essen, Germany
| | - Smiths S Lueong
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, 45122, Essen, Germany
| | - Jan Forster
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Department of Human Genetics, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Susanne Horn
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Marc Remke
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Ehrmann
- Center for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
- Department of Microbiology, University of Duisburg-Essen, Universitätsstraße 2, 45117, Essen, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Jürgen C Becker
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), University Hospital of Essen, Universitätsstrasse 1, 45141, Essen, Germany
| | - Iris Helfrich
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
| | - Daniel Rauh
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - Markus Kaiser
- Department of Chemical Biology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
- Center for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, 22525, Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Schnackenburgallee 114, 22525, Hamburg, Germany
| | | | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - José Neptuno Rodríguez-López
- GENZ-Group of Research on Enzymology, Department of Biochemistry and Molecular Biology-A, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Murcia, Spain
| | - Nikolas K Haass
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany
- Center for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen, Essen, Germany.
- German Consortium for Translational Cancer Research (DKTK), Partner Site Essen/Düsseldorf, Essen/Düsseldorf, Germany.
- Center for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
10
|
Guo S, Xue J, Liu J, Ye X, Guo Y, Liu D, Zhao X, Xiong F, Han X, Peng H. Smart imaging to empower brain-wide neuroscience at single-cell levels. Brain Inform 2022; 9:10. [PMID: 35543774 PMCID: PMC9095808 DOI: 10.1186/s40708-022-00158-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 11/10/2022] Open
Abstract
A deep understanding of the neuronal connectivity and networks with detailed cell typing across brain regions is necessary to unravel the mechanisms behind the emotional and memorial functions as well as to find the treatment of brain impairment. Brain-wide imaging with single-cell resolution provides unique advantages to access morphological features of a neuron and to investigate the connectivity of neuron networks, which has led to exciting discoveries over the past years based on animal models, such as rodents. Nonetheless, high-throughput systems are in urgent demand to support studies of neural morphologies at larger scale and more detailed level, as well as to enable research on non-human primates (NHP) and human brains. The advances in artificial intelligence (AI) and computational resources bring great opportunity to 'smart' imaging systems, i.e., to automate, speed up, optimize and upgrade the imaging systems with AI and computational strategies. In this light, we review the important computational techniques that can support smart systems in brain-wide imaging at single-cell resolution.
Collapse
Affiliation(s)
- Shuxia Guo
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China.
| | - Jie Xue
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Jian Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Xiangqiao Ye
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Yichen Guo
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Di Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Xuan Zhao
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Feng Xiong
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Xiaofeng Han
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China.
| | - Hanchuan Peng
- Institute for Brain and Intelligence, Southeast University, Nanjing, 210096, Jiangsu, China
| |
Collapse
|
11
|
Fei K, Zhang J, Yuan J, Xiao P. Present Application and Perspectives of Organoid Imaging Technology. Bioengineering (Basel) 2022; 9:121. [PMID: 35324810 PMCID: PMC8945799 DOI: 10.3390/bioengineering9030121] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/23/2022] [Accepted: 03/13/2022] [Indexed: 11/18/2022] Open
Abstract
An organoid is a miniaturized and simplified in vitro model with a similar structure and function to a real organ. In recent years, the use of organoids has increased explosively in the field of growth and development, disease simulation, drug screening, cell therapy, etc. In order to obtain necessary information, such as morphological structure, cell function and dynamic signals, it is necessary and important to directly monitor the culture process of organoids. Among different detection technologies, imaging technology is a simple and convenient choice and can realize direct observation and quantitative research. In this review, the principle, advantages and disadvantages of imaging technologies that have been applied in organoids research are introduced. We also offer an overview of prospective technologies for organoid imaging. This review aims to help biologists find appropriate imaging techniques for different areas of organoid research, and also contribute to the development of organoid imaging systems.
Collapse
Affiliation(s)
| | | | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China; (K.F.); (J.Z.)
| | - Peng Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China; (K.F.); (J.Z.)
| |
Collapse
|
12
|
Rosen ME, Dallon JC. A mathematical analysis of focal adhesion lifetimes and their effect on cell motility. Biophys J 2022; 121:1070-1080. [PMID: 35143774 PMCID: PMC8943753 DOI: 10.1016/j.bpj.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/07/2021] [Accepted: 02/03/2022] [Indexed: 11/02/2022] Open
Abstract
By analyzing the distributions of focal adhesion (FA) lifetimes from different cell types, we found that a gamma distribution best matched the experimental distributions. In all but one case, it was a unimodal, non-symmetric gamma distribution. We used a mathematical model of cell motion to help understand the mechanics and data behind the FA lifetime distributions. The model uses a detach-rate function to determine how long an FA will persist before it detaches. The detach-rate function that produced distributions with a best-fit gamma curve that closely matched that of the data was both force and time dependent. Using the data gathered from the matching simulations, we calculated both the cell speed and mean FA lifetime and compared them. Where available, we also compared this relationship to that of the experimental data and found that the simulation reasonably matches it in most cases. In both the simulations and experimental data, the cell speed and mean FA lifetime are related, with longer mean lifetimes being indicative of slower speeds. We suspect that one of the main predictors of cell speed for migrating cells is the distribution of the FA lifetimes.
Collapse
Affiliation(s)
- Mary Ellen Rosen
- Department of Mathematics, Brigham Young University, Provo, Utah
| | - J C Dallon
- Department of Mathematics, Brigham Young University, Provo, Utah.
| |
Collapse
|
13
|
Dema A, van Haren J, Wittmann T. Optogenetic EB1 inactivation shortens metaphase spindles by disrupting cortical force-producing interactions with astral microtubules. Curr Biol 2022; 32:1197-1205.e4. [PMID: 35090591 PMCID: PMC8930524 DOI: 10.1016/j.cub.2022.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/09/2021] [Accepted: 01/07/2022] [Indexed: 10/19/2022]
Abstract
Chromosome segregation is accomplished by the mitotic spindle, a bipolar micromachine built primarily from microtubules. Different microtubule populations contribute to spindle function: kinetochore microtubules attach and transmit forces to chromosomes, antiparallel interpolar microtubules support spindle structure, and astral microtubules connect spindle poles to the cell cortex.1,2 In mammalian cells, end-binding (EB) proteins associate with all growing microtubule plus ends throughout the cell cycle and serve as adaptors for diverse +TIPs that control microtubule dynamics and interactions with other intracellular structures.3 Because binding of many +TIPs to EB1 and thus microtubule-end association is switched off by mitotic phosphorylation,4-6 the mitotic function of EBs remains poorly understood. To analyze how EB1 and associated +TIPs on different spindle microtubule populations contribute to mitotic spindle dynamics, we use a light-sensitive EB1 variant, π-EB1, that allows local, acute, and reversible inactivation of +TIP association with growing microtubule ends in live cells.7 We find that acute π-EB1 photoinactivation results in rapid and reversible metaphase spindle shortening and transient relaxation of tension across the central spindle. However, in contrast to interphase, π-EB1 photoinactivation does not inhibit microtubule growth in metaphase but instead increases astral microtubule length and number. Yet in the absence of EB1 activity, astral microtubules fail to engage the cortical dynein/dynactin machinery, and spindle poles move away from regions of π-EB1 photoinactivation. In conclusion, our optogenetic approach reveals mitotic EB1 functions that remain hidden in genetic experiments, likely due to compensatory molecular systems regulating vertebrate spindle dynamics.
Collapse
Affiliation(s)
- Alessandro Dema
- Department of Cell & Tissue Biology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | | | - Torsten Wittmann
- Department of Cell & Tissue Biology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
14
|
Ge B, Zhang Q, Zhang R, Lin JT, Tseng PH, Chang CW, Dong CY, Zhou R, Yaqoob Z, Bischofberger I, So PTC. Single-Shot Quantitative Polarization Imaging of Complex Birefringent Structure Dynamics. ACS PHOTONICS 2021; 8:3440-3447. [PMID: 37292495 PMCID: PMC10249439 DOI: 10.1021/acsphotonics.1c00788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Polarization light microscopes are powerful tools for probing molecular order and orientation in birefringent materials. While a number of polarization microscopy techniques are available to access steady-state properties of birefringent samples, quantitative measurements of the molecular orientation dynamics on the millisecond time scale have remained a challenge. We propose polarized shearing interference microscopy (PSIM), a single-shot quantitative polarization imaging method, for extracting the retardance and orientation angle of the laser beam transmitting through optically anisotropic specimens with complex structures. The measurement accuracy and imaging performance of PSIM are validated by imaging a birefringent resolution target and a bovine tendon specimen. We demonstrate that PSIM can quantify the dynamics of a flowing lyotropic chromonic liquid crystal in a microfluidic channel at an imaging speed of 506 frames per second (only limited by the camera frame rate), with a field-of-view of up to 350 × 350 μm2 and a diffraction-limit spatial resolution of ~2 μm. We envision that PSIM will find a broad range of applications in quantitative material characterization under dynamical conditions.
Collapse
Affiliation(s)
- Baoliang Ge
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Qing Zhang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Rui Zhang
- Department of Physics, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Jing-Tang Lin
- Department of Physics, National Taiwan University, Taipei 106 Taiwan, Republic of China
| | - Po-Hang Tseng
- Department of Physics, National Taiwan University, Taipei 106 Taiwan, Republic of China
| | - Che-Wei Chang
- Department of Physics, National Taiwan University, Taipei 106 Taiwan, Republic of China
| | - Chen-Yuan Dong
- Department of Physics, National Taiwan University, Taipei 106 Taiwan, Republic of China
| | - Renjie Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, New Territories, Hong Kong 999077, China
| | - Zahid Yaqoob
- Laser Biomedical Research Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Irmgard Bischofberger
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Peter T C So
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Laser Biomedical Research Center and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Janssen BGH, Najiminaini M, Zhang YM, Omidi P, Carson JJL. Multispectral intravital microscopy for simultaneous bright-field and fluorescence imaging of the microvasculature. Appl Microsc 2021; 51:12. [PMID: 34302534 PMCID: PMC8310548 DOI: 10.1186/s42649-021-00059-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/08/2021] [Indexed: 12/05/2022] Open
Abstract
Intravital video microscopy permits the observation of microcirculatory blood flow. This often requires fluorescent probes to visualize structures and dynamic processes that cannot be observed with conventional bright-field microscopy. Conventional light microscopes do not allow for simultaneous bright-field and fluorescent imaging. Moreover, in conventional microscopes, only one type of fluorescent label can be observed. This study introduces multispectral intravital video microscopy, which combines bright-field and fluorescence microscopy in a standard light microscope. The technique enables simultaneous real-time observation of fluorescently-labeled structures in relation to their direct physical surroundings. The advancement provides context for the orientation, movement, and function of labeled structures in the microcirculation.
Collapse
Affiliation(s)
- Barry G H Janssen
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada. .,Kidney Clinical Research Unit (KCRU), London Health Sciences Centre, London, ON, N6C 6B5, Canada.
| | - Mohamadreza Najiminaini
- Imaging Program, St.Joseph's Health Care, Lawson Health Research Institute, London, ON, N6A 4V2, Canada.,Department of Pathology, Western University, London, ON, N6A 5C1, Canada
| | - Yan Min Zhang
- Kidney Clinical Research Unit (KCRU), London Health Sciences Centre, London, ON, N6C 6B5, Canada.,Trauma Research Centre, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100048, People's Republic of China.,School of Biomedical Engineering, Western University, London, ON, N6A 3K7, Canada.,Intensive Care Unit, Tianjin Nankai Hospital, Tianjin, 300100, People's Republic of China
| | - Parsa Omidi
- Imaging Program, St.Joseph's Health Care, Lawson Health Research Institute, London, ON, N6A 4V2, Canada.,Department of Pathology, Western University, London, ON, N6A 5C1, Canada.,Intensive Care Unit, Tianjin Nankai Hospital, Tianjin, 300100, People's Republic of China
| | - Jeffrey J L Carson
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada.,Imaging Program, St.Joseph's Health Care, Lawson Health Research Institute, London, ON, N6A 4V2, Canada.,Department of Pathology, Western University, London, ON, N6A 5C1, Canada.,Intensive Care Unit, Tianjin Nankai Hospital, Tianjin, 300100, People's Republic of China
| |
Collapse
|
16
|
Cordelli E, Soda P, Iannello G. Visual4DTracker: a tool to interact with 3D + t image stacks. BMC Bioinformatics 2021; 22:53. [PMID: 33557754 PMCID: PMC7869512 DOI: 10.1186/s12859-020-03820-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 10/15/2020] [Indexed: 11/29/2022] Open
Abstract
Background Biological phenomena usually evolves over time and recent advances in high-throughput microscopy have made possible to collect multiple 3D images over time, generating \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$3D+t$$\end{document}3D+t (or 4D) datasets. To extract useful information there is the need to extract spatial and temporal data on the particles that are in the images, but particle tracking and feature extraction need some kind of assistance. Results This manuscript introduces our new freely downloadable toolbox, the Visual4DTracker. It is a MATLAB package implementing several useful functionalities to navigate, analyse and proof-read the track of each particle detected in any \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$3D+t$$\end{document}3D+t stack. Furthermore, it allows users to proof-read and to evaluate the traces with respect to a given gold standard. The Visual4DTracker toolbox permits the users to visualize and save all the generated results through a user-friendly graphical user interface. This tool has been successfully used in three applicative examples. The first processes synthetic data to show all the software functionalities. The second shows how to process a 4D image stack showing the time-lapse growth of Drosophila cells in an embryo. The third example presents the quantitative analysis of insulin granules in living beta-cells, showing that such particles have two main dynamics that coexist inside the cells. Conclusions Visual4DTracker is a software package for MATLAB to visualize, handle and manually track \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$3D+t$$\end{document}3D+t stacks of microscopy images containing objects such cells, granules, etc.. With its unique set of functions, it remarkably permits the user to analyze and proof-read 4D data in a friendly 3D fashion. The tool is freely available at https://drive.google.com/drive/folders/19AEn0TqP-2B8Z10kOavEAopTUxsKUV73?usp=sharing
Collapse
Affiliation(s)
- Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy.
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Giulio Iannello
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
17
|
Zhang J, Velmeshev D, Hashimoto K, Huang YH, Hofmann JW, Shi X, Chen J, Leidal AM, Dishart JG, Cahill MK, Kelley KW, Liddelow SA, Seeley WW, Miller BL, Walther TC, Farese RV, Taylor JP, Ullian EM, Huang B, Debnath J, Wittmann T, Kriegstein AR, Huang EJ. Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency. Nature 2020; 588:459-465. [PMID: 32866962 PMCID: PMC7746606 DOI: 10.1038/s41586-020-2709-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/21/2020] [Indexed: 12/21/2022]
Abstract
Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.
Collapse
Affiliation(s)
- Jiasheng Zhang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Pathology Service 113B, San Francisco VA Health Care System, San Francisco, CA, USA
| | - Dmitry Velmeshev
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Kei Hashimoto
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Yu-Hsin Huang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey W Hofmann
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Xiaoyu Shi
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Jiapei Chen
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Andrew M Leidal
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Julian G Dishart
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Michelle K Cahill
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Kevin W Kelley
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Shane A Liddelow
- Neuroscience Institute, Department of Neuroscience & Physiology, NYU Langone Medical Center, New York, NY, USA
| | - William W Seeley
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Tobias C Walther
- Department of Genetics and Complex Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Robert V Farese
- Department of Genetics and Complex Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St Jude Children's Hospital & Howard Hughes Medical Institute, Memphis, TN, USA
| | - Erik M Ullian
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jayanta Debnath
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Torsten Wittmann
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
- Pathology Service 113B, San Francisco VA Health Care System, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Webb BA, Aloisio FM, Charafeddine RA, Cook J, Wittmann T, Barber DL. pHLARE: a new biosensor reveals decreased lysosome pH in cancer cells. Mol Biol Cell 2020; 32:131-142. [PMID: 33237838 PMCID: PMC8120692 DOI: 10.1091/mbc.e20-06-0383] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many lysosome functions are determined by a lumenal pH of ∼5.0, including the activity of resident acid-activated hydrolases. Lysosome pH (pHlys) is often increased in neurodegenerative disorders and predicted to be decreased in cancers, making it a potential target for therapeutics to limit the progression of these diseases. Accurately measuring pHlys, however, is limited by currently used dyes that accumulate in multiple intracellular compartments and cannot be propagated in clonal cells for longitudinal studies or used for in vivo determinations. To resolve this limitation, we developed a genetically encoded ratiometric pHlys biosensor, pHLARE (pHLysosomal Activity REporter), which localizes predominantly in lysosomes, has a dynamic range of pH 4.0 to 6.5, and can be stably expressed in cells. Using pHLARE we show decreased pHlys with inhibiting activity of the mammalian target of rapamycin complex 1 (mTORC1). Also, cancer cells from different tissue origins have a lower pHlys than untransformed cells, and stably expressing oncogenic RasV12 in untransformed cells is sufficient to decrease pHlys. pHLARE is a new tool to accurately measure pHlys for improved understanding of lysosome dynamics, which is increasingly considered a therapeutic target.
Collapse
Affiliation(s)
- Bradley A Webb
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94941
| | - Francesca M Aloisio
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94941
| | - Rabab A Charafeddine
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94941
| | - Jessica Cook
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94941
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94941
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94941
| |
Collapse
|
19
|
Luo F, Qin G, Xia T, Fang X. Single-Molecule Imaging of Protein Interactions and Dynamics. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:337-361. [PMID: 32228033 DOI: 10.1146/annurev-anchem-091619-094308] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Live-cell single-molecule fluorescence imaging has become a powerful analytical tool to investigate cellular processes that are not accessible to conventional biochemical approaches. This has greatly enriched our understanding of the behaviors of single biomolecules in their native environments and their roles in cellular events. Here, we review recent advances in fluorescence-based single-molecule bioimaging of proteins in living cells. We begin with practical considerations of the design of single-molecule fluorescence imaging experiments such as the choice of imaging modalities, fluorescent probes, and labeling methods. We then describe analytical observables from single-molecule data and the associated molecular parameters along with examples of live-cell single-molecule studies. Lastly, we discuss computational algorithms developed for single-molecule data analysis.
Collapse
Affiliation(s)
- Fang Luo
- Beijing National Research Center for Molecular Sciences, CAS Key Laboratory of Molecule Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China;
- Department of Chemistry, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Gege Qin
- Beijing National Research Center for Molecular Sciences, CAS Key Laboratory of Molecule Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China;
- Department of Chemistry, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Tie Xia
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaohong Fang
- Beijing National Research Center for Molecular Sciences, CAS Key Laboratory of Molecule Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China;
- Department of Chemistry, University of the Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Teo JL, Gomez GA, Weeratunga S, Davies EM, Noordstra I, Budnar S, Katsuno-Kambe H, McGrath MJ, Verma S, Tomatis V, Acharya BR, Balasubramaniam L, Templin RM, McMahon KA, Lee YS, Ju RJ, Stebhens SJ, Ladoux B, Mitchell CA, Collins BM, Parton RG, Yap AS. Caveolae Control Contractile Tension for Epithelia to Eliminate Tumor Cells. Dev Cell 2020; 54:75-91.e7. [PMID: 32485139 DOI: 10.1016/j.devcel.2020.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/17/2020] [Accepted: 05/01/2020] [Indexed: 01/24/2023]
Abstract
Epithelia are active materials where mechanical tension governs morphogenesis and homeostasis. But how that tension is regulated remains incompletely understood. We now report that caveolae control epithelial tension and show that this is necessary for oncogene-transfected cells to be eliminated by apical extrusion. Depletion of caveolin-1 (CAV1) increased steady-state tensile stresses in epithelial monolayers. As a result, loss of CAV1 in the epithelial cells surrounding oncogene-expressing cells prevented their apical extrusion. Epithelial tension in CAV1-depleted monolayers was increased by cortical contractility at adherens junctions. This reflected a signaling pathway, where elevated levels of phosphoinositide-4,5-bisphosphate (PtdIns(4,5)P2) recruited the formin, FMNL2, to promote F-actin bundling. Steady-state monolayer tension and oncogenic extrusion were restored to CAV1-depleted monolayers when tension was corrected by depleting FMNL2, blocking PtdIns(4,5)P2, or disabling the interaction between FMNL2 and PtdIns(4,5)P2. Thus, caveolae can regulate active mechanical tension for epithelial homeostasis by controlling lipid signaling to the actin cytoskeleton.
Collapse
Affiliation(s)
- Jessica L Teo
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Guillermo A Gomez
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia
| | - Saroja Weeratunga
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Elizabeth M Davies
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Ivar Noordstra
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Srikanth Budnar
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Hiroko Katsuno-Kambe
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Meagan J McGrath
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Suzie Verma
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Vanesa Tomatis
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Bipul R Acharya
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | | | - Rachel M Templin
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Kerrie-Ann McMahon
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Yoke Seng Lee
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Robert J Ju
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Samantha J Stebhens
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Benoit Ladoux
- Institut Jacques Monod, Université de Paris, CNRS UMR 7592, 75013 Paris, France
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Brett M Collins
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia
| | - Robert G Parton
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia.
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
21
|
Carceles-Cordon M, Kelly WK, Gomella L, Knudsen KE, Rodriguez-Bravo V, Domingo-Domenech J. Cellular rewiring in lethal prostate cancer: the architect of drug resistance. Nat Rev Urol 2020; 17:292-307. [PMID: 32203305 PMCID: PMC7218925 DOI: 10.1038/s41585-020-0298-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Over the past 5 years, the advent of combination therapeutic strategies has substantially reshaped the clinical management of patients with advanced prostate cancer. However, most of these combination regimens were developed empirically and, despite offering survival benefits, are not enough to halt disease progression. Thus, the development of effective therapeutic strategies that target the mechanisms involved in the acquisition of drug resistance and improve clinical trial design are an unmet clinical need. In this context, we hypothesize that the tumour engineers a dynamic response through the process of cellular rewiring, in which it adapts to the therapy used and develops mechanisms of drug resistance via downstream signalling of key regulatory cascades such as the androgen receptor, PI3K-AKT or GATA2-dependent pathways, as well as initiation of biological processes to revert tumour cells to undifferentiated aggressive states via phenotype switching towards a neuroendocrine phenotype or acquisition of stem-like properties. These dynamic responses are specific for each patient and could be responsible for treatment failure despite multi-target approaches. Understanding the common stages of these cellular rewiring mechanisms to gain a new perspective on the molecular underpinnings of drug resistance might help formulate novel combination therapeutic regimens.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - W Kevin Kelly
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard Gomella
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karen E Knudsen
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Veronica Rodriguez-Bravo
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Josep Domingo-Domenech
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Abstract
Light can be controlled with high spatial and temporal accuracy. Therefore, optogenetics is an attractive experimental approach to modulate intracellular cytoskeleton dynamics at much faster timescales than by genetic modification. For example, in mammalian cells, microtubules (MTs) grow tens of micrometers per minute and many intracellular MT functions are mediated by a complex of +TIP proteins that dynamically associate with growing MT plus ends. EB1 is a central component of this +TIP protein network, and we recently developed a photo-inactivated π-EB1 by inserting a blue light-sensitive LOV2/Zdk1 module between the EB1 MT-binding domain and the +TIP adaptor domain. Blue light-induced π-EB1 photodissociation results in disassembly of the +TIP complex and strongly attenuates MT growth in mammalian cells.In this chapter, we discuss theoretical and practical aspects of how to perform high-resolution live-cell microscopy in combination with π-EB1 photodissociation. However, these techniques are broadly applicable to other LOV2-based and likely other blue light-sensitive optogenetics. In addition to being a tool to investigate +TIP functions acutely and with subcellular resolution, because of its dramatic and rapid change in intracellular localization, π-EB1 can serve as a powerful tool to test and characterize optogenetic illumination setups. We describe protocols on how to achieve micrometer-scale intracellular control of π-EB1 activity using patterned illumination, and we introduce a do-it-yourself LED cube design compatible with transmitted light microscopy in multiwell plates.
Collapse
Affiliation(s)
- Jeffrey van Haren
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA.,Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Lauren S Adachi
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
23
|
Kim CS, Kim W, Lee K, Yoo H. High-speed color three-dimensional measurement based on parallel confocal detection with a focus tunable lens. OPTICS EXPRESS 2019; 27:28466-28479. [PMID: 31684598 DOI: 10.1364/oe.27.028466] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/07/2019] [Indexed: 05/28/2023]
Abstract
Reflectance confocal microscopy is a widely used optical imaging technique for non-destructive three-dimensional (3D) surface measurement. In confocal microscopy, a stack of two-dimensional (2D) images along the axial position is used for 3D reconstruction. This means the speed of 3D volumetric acquisition is limited by the beam scanning and the mechanical axial scanning. To achieve fast volumetric imaging, simultaneous multiple point scanning by parallelizing the beam instead of transverse point scanning can be considered, using a pinhole array. Previously, we developed a direct-view confocal microscope with a focus tunable lens (FTL) to produce a monochrome 3D surface profile of a sample without any mechanical scanning. Here, we report a high-speed color 3D measurement method based on parallel confocal detection. The proposed method produces a color 3D image of an object by acquiring 180 2D color images with an acquisition time of 1 second. We also visualized the color information of the object by overlaying the color obtained with a color area detector and a white LED illumination on top of the 3D surface profile. In addition, we designed an improved optical system to reduce artifacts caused by internal reflections and developed a new algorithm for noise-resistant 3D measurements. The feasibility of the proposed non-contact high-speed color 3D measurement for use in industrial or biomedical fields was demonstrated by imaging the color 3D shapes of various specimens. We anticipate that this technology can be utilized in various fields, where rapid 3D surface profiles with color information are required.
Collapse
|
24
|
Chong A, Starr T, Finn CE, Steele-Mortimer O. A role for the Salmonella Type III Secretion System 1 in bacterial adaptation to the cytosol of epithelial cells. Mol Microbiol 2019; 112:1270-1283. [PMID: 31370104 DOI: 10.1111/mmi.14361] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2019] [Indexed: 12/15/2022]
Abstract
Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that invades the intestinal epithelium. Following invasion of epithelial cells, Salmonella survives and replicates within two distinct intracellular niches. While all of the bacteria are initially taken up into a membrane bound vacuole, the Salmonella-containing vacuole or SCV, a significant proportion of them promptly escape into the cytosol. Cytosolic Salmonella replicates more rapidly compared to the vacuolar population, although the reasons for this are not well understood. SipA, a multi-function effector protein, has been shown to affect intracellular replication and is secreted by cytosolic Salmonella via the invasion-associated Type III Secretion System 1 (T3SS1). Here, we have used a multipronged microscopy approach to show that SipA does not affect bacterial replication rates per se, but rather mediates intra-cytosolic survival and/or initiation of replication following bacterial egress from the SCV. Altogether, our findings reveal an important role for SipA in the early survival of cytosolic Salmonella.
Collapse
Affiliation(s)
- Audrey Chong
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Tregei Starr
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ciaran E Finn
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Olivia Steele-Mortimer
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
25
|
Charafeddine RA, Cortopassi WA, Lak P, Tan R, McKenney RJ, Jacobson MP, Barber DL, Wittmann T. Tau repeat regions contain conserved histidine residues that modulate microtubule-binding in response to changes in pH. J Biol Chem 2019; 294:8779-8790. [PMID: 30992364 DOI: 10.1074/jbc.ra118.007004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/13/2019] [Indexed: 12/20/2022] Open
Abstract
Tau, a member of the MAP2/tau family of microtubule-associated proteins, stabilizes and organizes axonal microtubules in healthy neurons. In neurodegenerative tauopathies, tau dissociates from microtubules and forms neurotoxic extracellular aggregates. MAP2/tau family proteins are characterized by three to five conserved, intrinsically disordered repeat regions that mediate electrostatic interactions with the microtubule surface. Here, we used molecular dynamics, microtubule-binding experiments, and live-cell microscopy, revealing that highly-conserved histidine residues near the C terminus of each microtubule-binding repeat are pH sensors that can modulate tau-microtubule interaction strength within the physiological intracellular pH range. We observed that at low pH (<7.5), these histidines are positively charged and interact with phenylalanine residues in a hydrophobic cleft between adjacent tubulin dimers. At higher pH (>7.5), tau deprotonation decreased binding to microtubules both in vitro and in cells. Electrostatic and hydrophobic characteristics of histidine were both required for tau-microtubule binding, as substitutions with constitutively and positively charged nonaromatic lysine or uncharged alanine greatly reduced or abolished tau-microtubule binding. Consistent with these findings, tau-microtubule binding was reduced in a cancer cell model with increased intracellular pH but was rapidly restored by decreasing the pH to normal levels. These results add detailed insights into the intracellular regulation of tau activity that may be relevant in both normal and pathological conditions.
Collapse
Affiliation(s)
- Rabab A Charafeddine
- From the Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California 94143
| | - Wilian A Cortopassi
- the Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, and
| | - Parnian Lak
- the Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, and
| | - Ruensern Tan
- the Department of Molecular and Cellular Biology, University of California Davis, Davis, California 95616
| | - Richard J McKenney
- the Department of Molecular and Cellular Biology, University of California Davis, Davis, California 95616
| | - Matthew P Jacobson
- the Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, and
| | - Diane L Barber
- From the Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California 94143
| | - Torsten Wittmann
- From the Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, California 94143,
| |
Collapse
|
26
|
Packer LM, Stehbens SJ, Bonazzi VF, Gunter JH, Ju RJ, Ward M, Gartside MG, Byron SA, Pollock PM. Bcl-2 inhibitors enhance FGFR inhibitor-induced mitochondrial-dependent cell death in FGFR2-mutant endometrial cancer. Mol Oncol 2019; 13:738-756. [PMID: 30537101 PMCID: PMC6441928 DOI: 10.1002/1878-0261.12422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/10/2018] [Accepted: 11/19/2018] [Indexed: 01/09/2023] Open
Abstract
Endometrial cancer is the most commonly diagnosed gynaecological malignancy. Unfortunately, 15–20% of women demonstrate persistent or recurrent tumours that are refractory to current chemotherapies. We previously identified activating mutations in fibroblast growth factor receptor 2 (FGFR2) in 12% (stage I/II) to 17% (stage III/IV) endometrioid ECs and found that these mutations are associated with shorter progression‐free and cancer‐specific survival. Although FGFR inhibitors are undergoing clinical trials for treatment of several cancer types, little is known about the mechanism by which they induce cell death. We show that treatment with BGJ398, AZD4547 and PD173074 causes mitochondrial depolarization, cytochrome c release and impaired mitochondrial respiration in two FGFR2‐mutant EC cell lines (AN3CA and JHUEM2). Despite this mitochondrial dysfunction, we were unable to detect caspase activation following FGFR inhibition; in addition, the pan‐caspase inhibitor Z‐VAD‐FMK was unable to prevent cell death, suggesting that the cell death is caspase‐independent. Furthermore, while FGFR inhibition led to an increase in LC3 puncta, treatment with bafilomycin did not further increase lipidated LC3, suggesting that FGFR inhibition led to a block in autophagosome degradation. We confirmed that cell death is mitochondrial‐dependent as it can be blocked by overexpression of Bcl‐2 and/or Bcl‐XL. Importantly, we show that combining FGFR inhibitors with the BH3 mimetics ABT737/ABT263 markedly increased cell death in vitro and is more effective than BGJ398 alone in vivo, where it leads to marked tumour regression. This work may have implications for the design of clinical trials to treat a wide range of patients with FGFR‐dependent malignancies.
Collapse
Affiliation(s)
- Leisl M Packer
- School of Biomedical Science, Institute of Health & Biomedical Innovation, Queensland University of Technology located within the Translational Research Institute, Brisbane, Australia
| | - Samantha J Stehbens
- School of Biomedical Science, Institute of Health & Biomedical Innovation, Queensland University of Technology located within the Translational Research Institute, Brisbane, Australia
| | - Vanessa F Bonazzi
- School of Biomedical Science, Institute of Health & Biomedical Innovation, Queensland University of Technology located within the Translational Research Institute, Brisbane, Australia
| | - Jennifer H Gunter
- School of Biomedical Science, Institute of Health & Biomedical Innovation, Queensland University of Technology located within the Translational Research Institute, Brisbane, Australia
| | - Robert J Ju
- School of Biomedical Science, Institute of Health & Biomedical Innovation, Queensland University of Technology located within the Translational Research Institute, Brisbane, Australia
| | - Micheal Ward
- Mater-UQ located within the Translational Research Institute, Brisbane, Australia
| | - Michael G Gartside
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Sara A Byron
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Pamela M Pollock
- School of Biomedical Science, Institute of Health & Biomedical Innovation, Queensland University of Technology located within the Translational Research Institute, Brisbane, Australia
| |
Collapse
|
27
|
Turkowyd B, Müller-Esparza H, Climenti V, Steube N, Endesfelder U, Randau L. Live-cell single-particle tracking photoactivated localization microscopy of Cascade-mediated DNA surveillance. Methods Enzymol 2019; 616:133-171. [PMID: 30691641 DOI: 10.1016/bs.mie.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Type I CRISPR-Cas systems utilize small CRISPR RNA (crRNA) molecules to scan DNA strands for target regions. Different crRNAs are bound by several CRISPR-associated (Cas) protein subunits that form the stable ribonucleoprotein complex Cascade. The Cascade-mediated DNA surveillance process requires a sufficient degree of base-complementarity between crRNA and target sequences and relies on the recognition of small DNA motifs, termed protospacer adjacent motifs. Recently, super-resolution microscopy and single-particle tracking methods have been developed to follow individual protein complexes in live cells. Here, we described how this technology can be adapted to visualize the DNA scanning process of Cascade assemblies in Escherichia coli cells. The activity of recombinant Type I-Fv Cascade complexes of Shewanella putrefaciens CN-32 serves as a model system that facilitates comparative studies for many of the diverse CRISPR-Cas systems.
Collapse
Affiliation(s)
- Bartosz Turkowyd
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany; LOEWE Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Hanna Müller-Esparza
- Prokaryotic Small RNA Biology Group, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Vanessa Climenti
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany; LOEWE Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Niklas Steube
- Prokaryotic Small RNA Biology Group, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Ulrike Endesfelder
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany; LOEWE Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany.
| | - Lennart Randau
- LOEWE Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany; Prokaryotic Small RNA Biology Group, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany.
| |
Collapse
|
28
|
Neagu AN. Proteome Imaging: From Classic to Modern Mass Spectrometry-Based Molecular Histology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:55-98. [PMID: 31347042 DOI: 10.1007/978-3-030-15950-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In order to overcome the limitations of classic imaging in Histology during the actually era of multiomics, the multi-color "molecular microscope" by its emerging "molecular pictures" offers quantitative and spatial information about thousands of molecular profiles without labeling of potential targets. Healthy and diseased human tissues, as well as those of diverse invertebrate and vertebrate animal models, including genetically engineered species and cultured cells, can be easily analyzed by histology-directed MALDI imaging mass spectrometry. The aims of this review are to discuss a range of proteomic information emerging from MALDI mass spectrometry imaging comparative to classic histology, histochemistry and immunohistochemistry, with applications in biology and medicine, concerning the detection and distribution of structural proteins and biological active molecules, such as antimicrobial peptides and proteins, allergens, neurotransmitters and hormones, enzymes, growth factors, toxins and others. The molecular imaging is very well suited for discovery and validation of candidate protein biomarkers in neuroproteomics, oncoproteomics, aging and age-related diseases, parasitoproteomics, forensic, and ecotoxicology. Additionally, in situ proteome imaging may help to elucidate the physiological and pathological mechanisms involved in developmental biology, reproductive research, amyloidogenesis, tumorigenesis, wound healing, neural network regeneration, matrix mineralization, apoptosis and oxidative stress, pain tolerance, cell cycle and transformation under oncogenic stress, tumor heterogeneity, behavior and aggressiveness, drugs bioaccumulation and biotransformation, organism's reaction against environmental penetrating xenobiotics, immune signaling, assessment of integrity and functionality of tissue barriers, behavioral biology, and molecular origins of diseases. MALDI MSI is certainly a valuable tool for personalized medicine and "Eco-Evo-Devo" integrative biology in the current context of global environmental challenges.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Iasi, Romania.
| |
Collapse
|
29
|
White KA, Grillo-Hill BK, Esquivel M, Peralta J, Bui VN, Chire I, Barber DL. β-Catenin is a pH sensor with decreased stability at higher intracellular pH. J Cell Biol 2018; 217:3965-3976. [PMID: 30315137 PMCID: PMC6219716 DOI: 10.1083/jcb.201712041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 07/16/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023] Open
Abstract
White et al. find that intracellular pH regulates the stability of β-catenin, the Wnt signaling molecule that controls cell polarity, adhesion, and differentiation. A conserved histidine residue in β-catenin mediates pH-dependent binding to the E3 ligase β-TrCP for degradation, and a cancer-associated mutation that bypasses this pH-sensitive regulation induces ectopic tumors in the Drosophila eye. β-Catenin functions as an adherens junction protein for cell–cell adhesion and as a signaling protein. β-catenin function is dependent on its stability, which is regulated by protein–protein interactions that stabilize β-catenin or target it for proteasome-mediated degradation. In this study, we show that β-catenin stability is regulated by intracellular pH (pHi) dynamics, with decreased stability at higher pHi in both mammalian cells and Drosophila melanogaster. β-Catenin degradation requires phosphorylation of N-terminal residues for recognition by the E3 ligase β-TrCP. While β-catenin phosphorylation was pH independent, higher pHi induced increased β-TrCP binding and decreased β-catenin stability. An evolutionarily conserved histidine in β-catenin (found in the β-TrCP DSGIHS destruction motif) is required for pH-dependent binding to β-TrCP. Expressing a cancer-associated H36R–β-catenin mutant in the Drosophila eye was sufficient to induce Wnt signaling and produced pronounced tumors not seen with other oncogenic β-catenin alleles. We identify pHi dynamics as a previously unrecognized regulator of β-catenin stability, functioning in coincidence with phosphorylation.
Collapse
Affiliation(s)
- Katharine A White
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| | - Bree K Grillo-Hill
- Department of Biological Sciences, San Jose State University, San Jose, CA
| | - Mario Esquivel
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| | - Jobelle Peralta
- Department of Biological Sciences, San Jose State University, San Jose, CA
| | - Vivian N Bui
- Department of Biological Sciences, San Jose State University, San Jose, CA
| | - Ismahan Chire
- Department of Biological Sciences, San Jose State University, San Jose, CA
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
30
|
Stehbens SJ, Ju RJ, Adams MN, Perry SR, Haass NK, Bryant DM, Pollock PM. FGFR2-activating mutations disrupt cell polarity to potentiate migration and invasion in endometrial cancer cell models. J Cell Sci 2018; 131:jcs.213678. [PMID: 30002137 DOI: 10.1242/jcs.213678] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/24/2018] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases that control a diverse range of biological processes during development and in adult tissues. We recently reported that somatic FGFR2 mutations are associated with shorter survival in endometrial cancer. However, little is known about how these FGFR2 mutations contribute to endometrial cancer metastasis. Here, we report that expression of the activating mutations FGFR2N550K and FGFR2Y376C in an endometrial cancer cell model induce Golgi fragmentation, and loss of polarity and directional migration. In mutant FGFR2-expressing cells, this was associated with an inability to polarise intracellular pools of FGFR2 towards the front of migrating cells. Such polarization defects were exacerbated in three-dimensional culture, where FGFR2 mutant cells were unable to form well-organised acini, instead undergoing exogenous ligand-independent invasion. Our findings uncover collective cell polarity and invasion as common targets of disease-associated FGFR2 mutations that lead to poor outcome in endometrial cancer patients.
Collapse
Affiliation(s)
- Samantha J Stehbens
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia .,The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Robert J Ju
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia.,The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Mark N Adams
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Samuel R Perry
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Nikolas K Haass
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - David M Bryant
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Pamela M Pollock
- School of Biomedical Sciences, Queensland University of Technology (QUT) located at the Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| |
Collapse
|
31
|
Rust K, Tiwari MD, Mishra VK, Grawe F, Wodarz A. Myc and the Tip60 chromatin remodeling complex control neuroblast maintenance and polarity in Drosophila. EMBO J 2018; 37:embj.201798659. [PMID: 29997178 DOI: 10.15252/embj.201798659] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 02/04/2023] Open
Abstract
Stem cells establish cortical polarity and divide asymmetrically to simultaneously maintain themselves and generate differentiating offspring cells. Several chromatin modifiers have been identified as stemness factors in mammalian pluripotent stem cells, but whether these factors control stem cell polarity and asymmetric division has not been investigated so far. We addressed this question in Drosophila neural stem cells called neuroblasts. We identified the Tip60 chromatin remodeling complex and its interaction partner Myc as regulators of genes required for neuroblast maintenance. Knockdown of Tip60 complex members results in loss of cortical polarity, symmetric neuroblast division, and premature differentiation through nuclear entry of the transcription factor Prospero. We found that aPKC is the key target gene of Myc and the Tip60 complex subunit Domino in regulating neuroblast polarity. Our transcriptome analysis further showed that Domino regulates the expression of mitotic spindle genes previously identified as direct Myc targets. Our findings reveal an evolutionarily conserved functional link between Myc, the Tip60 complex, and the molecular network controlling cell polarity and asymmetric cell division.
Collapse
Affiliation(s)
- Katja Rust
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Cologne, Germany .,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), Cologne, Germany.,Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Göttingen, Germany.,Department of Anatomy and OB-GYN/RS, University of California, San Francisco, San Francisco, CA, USA
| | - Manu D Tiwari
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Cologne, Germany.,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), Cologne, Germany.,Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Göttingen, Germany
| | - Vivek Kumar Mishra
- Department of Dermatology and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Ferdi Grawe
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Cologne, Germany
| | - Andreas Wodarz
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Cologne, Germany .,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), Cologne, Germany.,Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Göttingen, Germany
| |
Collapse
|
32
|
van Haren J, Charafeddine RA, Ettinger A, Wang H, Hahn KM, Wittmann T. Local control of intracellular microtubule dynamics by EB1 photodissociation. Nat Cell Biol 2018; 20:252-261. [PMID: 29379139 PMCID: PMC5826794 DOI: 10.1038/s41556-017-0028-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022]
Abstract
End-binding proteins, EBs, are adaptors that recruit functionally diverse +TIP proteins to growing microtubule plus ends. To test with high spatial and temporal accuracy how, when and where +TIP protein complexes contribute to dynamic cell biology, we developed a photo-inactivated EB1 variant (π-EB1) by inserting a blue light-sensitive protein-protein interaction module between the microtubule- and +TIP-binding domains of EB1. π-EB1 replaces endogenous EB1 function in the absence of blue light. In contrast, blue light-mediated π-EB1 photo-dissociation results in rapid +TIP complex disassembly, and acutely and reversibly attenuates microtubule growth independent of microtubule end association of the microtubule polymerase CKAP5 (ch-TOG, XMAP215). Local π-EB1 photo-dissociation allows subcellular microtubule dynamics control at the second and micrometre scale, and elicits aversive turning of migrating cancer cells. Importantly, light-mediated domain splitting can serve as template to optically control other intracellular protein activities.
Collapse
Affiliation(s)
- Jeffrey van Haren
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Rabab A Charafeddine
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Andreas Ettinger
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.,Institute of Epigenetics and Stem Cells, Helmholtz Center Munich, München, Germany
| | - Hui Wang
- University of North Carolina, Chapel Hill, NC, USA
| | - Klaus M Hahn
- University of North Carolina, Chapel Hill, NC, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.
| |
Collapse
|
33
|
Zhao R, Li N, Xu J, Li W, Fang X. Quantitative single-molecule study of TGF-β/Smad signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:51-59. [PMID: 29190315 DOI: 10.1093/abbs/gmx121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022] Open
Abstract
TGF-β/Smad signaling pathway triggers diverse cellular responses among different cell types and environmental conditions. Quantitative analysis of protein-protein interactions involved in TGF-β/Smad signaling is demanded for understanding the molecular mechanism of this signaling pathway. Live-cell single-molecule microcopy with high spatiotemporal resolution is a new tool to monitor key molecular events in a real-time manner. In this review, we mainly presented the recent work on the quantitative characterization of TGF-β/Smad signaling proteins by single-molecule method, and showed how it enabled us to obtain new insights about this canonical signaling process.
Collapse
Affiliation(s)
- Rong Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nan Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiachao Xu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenhui Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Fang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
34
|
O'Boyle N, Connolly JPR, Roe AJ. Tracking elusive cargo: Illuminating spatio-temporal Type 3 effector protein dynamics using reporters. Cell Microbiol 2017; 20. [PMID: 29087624 PMCID: PMC5765406 DOI: 10.1111/cmi.12797] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/13/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022]
Abstract
Type 3 secretion systems form an integral part of the arsenal of many pathogenic bacteria. These injection machines, together with their cargo of subversive effector proteins, are capable of manipulating the cellular environment of the host in order to ensure persistence of the pathogen. In order to fully appreciate the functions of Type 3 effectors, it is necessary to gain spatio‐temporal knowledge of each effector during the process of infection. A number of genetic modifications have been exploited in order to reveal effector protein secretion, translocation and subsequent activity, and localisation within host cells. In this review, we will discuss the many available approaches for tracking effector protein dynamics and discuss the challenges faced to improve the current technologies and gain a clearer picture of effector protein function.
Collapse
Affiliation(s)
- Nicky O'Boyle
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - James P R Connolly
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Andrew J Roe
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
35
|
Comprehensive analysis of normal adjacent to tumor transcriptomes. Nat Commun 2017; 8:1077. [PMID: 29057876 PMCID: PMC5651823 DOI: 10.1038/s41467-017-01027-z] [Citation(s) in RCA: 341] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022] Open
Abstract
Histologically normal tissue adjacent to the tumor (NAT) is commonly used as a control in cancer studies. However, little is known about the transcriptomic profile of NAT, how it is influenced by the tumor, and how the profile compares with non-tumor-bearing tissues. Here, we integrate data from the Genotype-Tissue Expression project and The Cancer Genome Atlas to comprehensively analyze the transcriptomes of healthy, NAT, and tumor tissues in 6506 samples across eight tissues and corresponding tumor types. Our analysis shows that NAT presents a unique intermediate state between healthy and tumor. Differential gene expression and protein–protein interaction analyses reveal altered pathways shared among NATs across tissue types. We characterize a set of 18 genes that are specifically activated in NATs. By applying pathway and tissue composition analyses, we suggest a pan-cancer mechanism of pro-inflammatory signals from the tumor stimulates an inflammatory response in the adjacent endothelium. Normal tissue adjacent to the tumour (NAT) is often used as a control in cancer studies. Here, the authors analyse across cancer types the transcriptomes of healthy, NAT, and tumour tissues, and find that NAT presents a unique state, potentially due to inflammatory response of the NAT to the tumour tissue.
Collapse
|
36
|
Imaging and lipidomics methods for lipid analysis in metabolic and cardiovascular disease. J Dev Orig Health Dis 2017; 8:566-574. [PMID: 28697812 DOI: 10.1017/s2040174417000496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiometabolic diseases exhibit changes in lipid biology, which is important as lipids have critical roles in membrane architecture, signalling, hormone synthesis, homoeostasis and metabolism. However, Developmental Origins of Health and Disease studies of cardiometabolic disease rarely include analysis of lipids. This short review highlights some examples of lipid pathology and then explores the technology available for analysing lipids, focussing on the need to develop imaging modalities for intracellular lipids. Analytical methods for studying interactions between the complex endocrine and intracellular signalling pathways that regulate lipid metabolism have been critical in expanding our understanding of how cardiometabolic diseases develop in association with obesity and dietary factors. Biochemical methods can be used to generate detailed lipid profiles to establish links between lifestyle factors and metabolic signalling pathways and determine how changes in specific lipid subtypes in plasma and homogenized tissue are associated with disease progression. New imaging modalities enable the specific visualization of intracellular lipid traffic and distribution in situ. These techniques provide a dynamic picture of the interactions between lipid storage, mobilization and signalling, which operate during normal cell function and are altered in many important diseases. The development of methods for imaging intracellular lipids can provide a dynamic real-time picture of how lipids are involved in complex signalling and other cell biology pathways; and how they ultimately regulate metabolic function/homoeostasis during early development. Some imaging modalities have the potential to be adapted for in vivo applications, and may enable the direct visualization of progression of pathogenesis of cardiometabolic disease after poor growth in early life.
Collapse
|
37
|
Li N, Zhao R, Sun Y, Ye Z, He K, Fang X. Single-molecule imaging and tracking of molecular dynamics in living cells. Natl Sci Rev 2017. [DOI: 10.1093/nsr/nww055] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
Unlike the ensemble-averaging measurements, the single-molecule imaging and tracking (SMIT) in living cells provides the real-time quantitative information about the locations, kinetics, dynamics and interactions of individual molecules in their native environments with high spatiotemporal resolution and minimal perturbation. The past decade has witnessed a transforming development in the methods of SMIT with living cells, including fluorescent probes, labeling strategies, fluorescence microscopy, and detection and tracking algorithms. In this review, we will discuss these aspects with a particular focus on their recent advancements. We will then describe representative single-molecule studies to illustrate how the single-molecule approaches can be applied to monitor biomolecular interaction/reaction dynamics, and extract the molecular mechanistic information for different cellular systems.
Collapse
Affiliation(s)
- Nan Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rong Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yahong Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zi Ye
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kangmin He
- Department of Cell Biology, Harvard Medical School, and Cellular and Molecular Medicine Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Xiaohong Fang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
38
|
Webb BA, Dosey AM, Wittmann T, Kollman JM, Barber DL. The glycolytic enzyme phosphofructokinase-1 assembles into filaments. J Cell Biol 2017. [PMID: 28646105 PMCID: PMC5551713 DOI: 10.1083/jcb.201701084] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Despite abundant knowledge of the regulation and biochemistry of glycolytic enzymes, we have limited understanding on how they are spatially organized in the cell. Emerging evidence indicates that nonglycolytic metabolic enzymes regulating diverse pathways can assemble into polymers. We now show tetramer- and substrate-dependent filament assembly by phosphofructokinase-1 (PFK1), which is considered the "gatekeeper" of glycolysis because it catalyzes the step committing glucose to breakdown. Recombinant liver PFK1 (PFKL) isoform, but not platelet PFK1 (PFKP) or muscle PFK1 (PFKM) isoforms, assembles into filaments. Negative-stain electron micrographs reveal that filaments are apolar and made of stacked tetramers oriented with exposed catalytic sites positioned along the edge of the polymer. Electron micrographs and biochemical data with a PFKL/PFKP chimera indicate that the PFKL regulatory domain mediates filament assembly. Quantified live-cell imaging shows dynamic properties of localized PFKL puncta that are enriched at the plasma membrane. These findings reveal a new behavior of a key glycolytic enzyme with insights on spatial organization and isoform-specific glucose metabolism in cells.
Collapse
Affiliation(s)
- Bradley A Webb
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| | - Anne M Dosey
- Department of Biochemistry, University of Washington, Seattle, WA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| | - Justin M Kollman
- Department of Biochemistry, University of Washington, Seattle, WA
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
39
|
Wagner M, Horn H. Optical coherence tomography in biofilm research: A comprehensive review. Biotechnol Bioeng 2017; 114:1386-1402. [DOI: 10.1002/bit.26283] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/10/2017] [Accepted: 03/01/2017] [Indexed: 01/29/2023]
Affiliation(s)
- Michael Wagner
- Karlsruhe Institute of Technology; Engler-Bunte-Institut; Chair of Water Chemistry and Water Technology; Engler-Bunte-Ring 9 76131 Karlsruhe Germany
- Karlsruhe Institute of Technology; Institute of Functional Interfaces; Eggenstein-Leopoldshafen Germany
| | - Harald Horn
- Karlsruhe Institute of Technology; Engler-Bunte-Institut; Chair of Water Chemistry and Water Technology; Engler-Bunte-Ring 9 76131 Karlsruhe Germany
| |
Collapse
|
40
|
Pemble H, Kumar P, van Haren J, Wittmann T. GSK3-mediated CLASP2 phosphorylation modulates kinetochore dynamics. J Cell Sci 2017; 130:1404-1412. [PMID: 28232523 DOI: 10.1242/jcs.194662] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 02/20/2017] [Indexed: 02/02/2023] Open
Abstract
Error-free chromosome segregation requires dynamic control of microtubule attachment to kinetochores, but how kinetochore-microtubule interactions are spatially and temporally controlled during mitosis remains incompletely understood. In addition to the NDC80 microtubule-binding complex, other proteins with demonstrated microtubule-binding activities localize to kinetochores. One such protein is the cytoplasmic linker-associated protein 2 (CLASP2). Here, we show that global GSK3-mediated phosphorylation of the longest isoform, CLASP2α, largely abolishes CLASP2α-microtubule association in metaphase. However, it does not directly control localization of CLASP2α to kinetochores. Using dominant phosphorylation-site variants, we find that CLASP2α phosphorylation weakens kinetochore-microtubule interactions as evidenced by decreased tension between sister kinetochores. Expression of CLASP2α phosphorylation-site mutants also resulted in increased chromosome segregation defects, indicating that GSK3-mediated control of CLASP2α-microtubule interactions contributes to correct chromosome dynamics. Because of global inhibition of CLASP2α-microtubule interactions, we propose a model in which only kinetochore-bound CLASP2α is dephosphorylated, locally engaging its microtubule-binding activity.
Collapse
Affiliation(s)
- Hayley Pemble
- Department of Cell & Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Praveen Kumar
- Department of Cell & Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jeffrey van Haren
- Department of Cell & Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Torsten Wittmann
- Department of Cell & Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| |
Collapse
|
41
|
Ettinger A, Kosodo Y, Huttner WB. Specific membrane dynamics during neural stem cell division. Methods Cell Biol 2016; 137:143-172. [PMID: 28065302 DOI: 10.1016/bs.mcb.2016.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Neural stem and progenitor cells in the developing cerebral cortex, but also when grown in culture, display a range of distinct phenomena during cytokinesis. Cleavage furrow ingression in neural progenitor cells can bisect their basal processes and, later on, result in midbody formation at the apical surface. After abscission, these midbodies are released as membrane-bound particles into the extracellular space, in contrast to uptake and degradation of postabscission midbodies in other cell types. Whether these cellular dynamics are unique to neural stem cells, or more ubiquitously found, and what biological significance these processes have for cell differentiation or cell-cell communication, are open questions that require a combination of approaches. Here, we discuss techniques to study the specific membrane dynamics underlying the basal process splitting and postabscission midbody release in neural stem cells. We provide some basic concepts and protocols to isolate, enrich and stain released midbodies, and follow midbody dynamics over time. Moreover, we discuss techniques to prepare cortical sections for high-voltage electron microscopy to visualize the fine basal processes of progenitor cells.
Collapse
Affiliation(s)
- A Ettinger
- Institute of Epigenetics and Stem Cells, Munich, Germany
| | - Y Kosodo
- Korea Brain Research Institute, Daegu, Korea
| | - W B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
42
|
Doublecortin Is Excluded from Growing Microtubule Ends and Recognizes the GDP-Microtubule Lattice. Curr Biol 2016; 26:1549-1555. [PMID: 27238282 DOI: 10.1016/j.cub.2016.04.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/03/2016] [Accepted: 04/08/2016] [Indexed: 12/14/2022]
Abstract
Many microtubule (MT) functions are mediated by a diverse class of proteins (+TIPs) at growing MT plus ends that control intracellular MT interactions and dynamics and depend on end-binding proteins (EBs) [1]. Cryoelectron microscopy has recently identified the EB binding site as the interface of four tubulin dimers that undergoes a conformational change in response to β-tubulin GTP hydrolysis [2, 3]. Doublecortin (DCX), a MT-associated protein (MAP) required for neuronal migration during cortical development [4, 5], binds to the same site as EBs [6], and recent in vitro studies proposed DCX localization to growing MT ends independent of EBs [7]. Because this conflicts with observations in neurons [8, 9] and the molecular function of DCX is not well understood, we revisited intracellular DCX dynamics at low expression levels. Here, we report that DCX is not a +TIP in cells but, on the contrary, is excluded from the EB1 domain. In addition, we find that DCX-MT interactions are highly sensitive to MT geometry. In cells, DCX binding was greatly reduced at MT segments with high local curvature. Remarkably, this geometry-dependent binding to MTs was completely reversed in the presence of taxanes, which reconciles incompatible observations in cells [9] and in vitro [10]. We propose a model explaining DCX specificity for different MT geometries based on structural changes induced by GTP hydrolysis that decreases the spacing between adjacent tubulin dimers [11]. Our data are consistent with a unique mode of MT interaction in which DCX specifically recognizes this compacted GDP-like MT lattice.
Collapse
|
43
|
Migration of Founder Epithelial Cells Drives Proper Molar Tooth Positioning and Morphogenesis. Dev Cell 2016; 35:713-24. [PMID: 26702830 DOI: 10.1016/j.devcel.2015.11.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/19/2015] [Accepted: 11/24/2015] [Indexed: 02/05/2023]
Abstract
The proper positioning of organs during development is essential, yet little is known about the regulation of this process in mammals. Using murine tooth development as a model, we have found that cell migration plays a central role in positioning of the organ primordium. By combining lineage tracing, genetic cell ablation, and confocal live imaging, we identified a migratory population of Fgf8-expressing epithelial cells in the embryonic mandible. These Fgf8-expressing progenitors furnish the epithelial cells required for tooth development, and the progenitor population migrates toward a Shh-expressing region in the mandible, where the tooth placode will initiate. Inhibition of Fgf and Shh signaling disrupted the oriented migration of cells, leading to a failure of tooth development. These results demonstrate the importance of intraepithelial cell migration in proper positioning of an initiating organ.
Collapse
|
44
|
|
45
|
Lane AB, Strzelecka M, Ettinger A, Grenfell AW, Wittmann T, Heald R. Enzymatically Generated CRISPR Libraries for Genome Labeling and Screening. Dev Cell 2015. [PMID: 26212133 DOI: 10.1016/j.devcel.2015.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
CRISPR-based technologies have emerged as powerful tools to alter genomes and mark chromosomal loci, but an inexpensive method for generating large numbers of RNA guides for whole genome screening and labeling is lacking. Using a method that permits library construction from any source of DNA, we generated guide libraries that label repetitive loci or a single chromosomal locus in Xenopus egg extracts and show that a complex library can target the E. coli genome at high frequency.
Collapse
Affiliation(s)
- Andrew B Lane
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720-3200, USA.
| | - Magdalena Strzelecka
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720-3200, USA
| | - Andreas Ettinger
- Department of Cell & Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0512, USA
| | - Andrew W Grenfell
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720-3200, USA
| | - Torsten Wittmann
- Department of Cell & Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0512, USA
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720-3200, USA.
| |
Collapse
|
46
|
Galler K, Bräutigam K, Große C, Popp J, Neugebauer U. Making a big thing of a small cell--recent advances in single cell analysis. Analyst 2015; 139:1237-73. [PMID: 24495980 DOI: 10.1039/c3an01939j] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Single cell analysis is an emerging field requiring a high level interdisciplinary collaboration to provide detailed insights into the complex organisation, function and heterogeneity of life. This review is addressed to life science researchers as well as researchers developing novel technologies. It covers all aspects of the characterisation of single cells (with a special focus on mammalian cells) from morphology to genetics and different omics-techniques to physiological, mechanical and electrical methods. In recent years, tremendous advances have been achieved in all fields of single cell analysis: (1) improved spatial and temporal resolution of imaging techniques to enable the tracking of single molecule dynamics within single cells; (2) increased throughput to reveal unexpected heterogeneity between different individual cells raising the question what characterizes a cell type and what is just natural biological variation; and (3) emerging multimodal approaches trying to bring together information from complementary techniques paving the way for a deeper understanding of the complexity of biological processes. This review also covers the first successful translations of single cell analysis methods to diagnostic applications in the field of tumour research (especially circulating tumour cells), regenerative medicine, drug discovery and immunology.
Collapse
Affiliation(s)
- Kerstin Galler
- Integrated Research and Treatment Center "Center for Sepsis Control and Care", Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| | | | | | | | | |
Collapse
|
47
|
Grillo-Hill BK, Choi C, Jimenez-Vidal M, Barber DL. Increased H⁺ efflux is sufficient to induce dysplasia and necessary for viability with oncogene expression. eLife 2015; 4. [PMID: 25793441 PMCID: PMC4392478 DOI: 10.7554/elife.03270] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Intracellular pH (pHi) dynamics is increasingly recognized as an important regulator of a range of normal and pathological cell behaviors. Notably, increased pHi is now acknowledged as a conserved characteristic of cancers and in cell models is confirmed to increase proliferation and migration as well as limit apoptosis. However, the significance of increased pHi for cancer in vivo remains unresolved. Using Drosophila melanogaster, we show that increased pHi is sufficient to induce dysplasia in the absence of other transforming cues and potentiates growth and invasion with oncogenic Ras. Using a genetically encoded biosensor we also confirm increased pHi in situ. Moreover, in Drosophila models and clonal human mammary cells we show that limiting H(+) efflux with oncogenic Raf or Ras induces acidosis and synthetic lethality. Further, we show lethality in invasive primary tumor cell lines with inhibiting H(+) efflux. Synthetic lethality with reduced H(+) efflux and activated oncogene expression could be exploited therapeutically to restrain cancer progression while limiting off-target effects.
Collapse
Affiliation(s)
- Bree K Grillo-Hill
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Changhoon Choi
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Maite Jimenez-Vidal
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
48
|
Abstract
During the intracellular phase of the pathogenic lifestyle, Salmonella enterica massively alters the endosomal system of its host cells. Two hallmarks are the remodeling of phagosomes into the Salmonella-containing vacuole (SCV) as a replicative niche, and the formation of tubular structures, such as Salmonella-induced filaments (SIFs). To study the dynamics and the fate of these Salmonella-specific compartments, live cell imaging (LCI) is a method of choice. In this chapter, we compare currently used microscopy techniques and focus on considerations and requirements specific for LCI. Detailed protocols for LCI of Salmonella infection with either confocal laser scanning microscopy (CLSM) or spinning disk confocal microscopy (SDCM) are provided.
Collapse
Affiliation(s)
- Alexander Kehl
- Abteilung Mikrobiologie, Fachbereich Biologie/Chemie, Universität Osnabrück, Barbarastr. 11, Osnabrück, 49076, Germany
| | | |
Collapse
|
49
|
Nedvetsky PI, Emmerson E, Finley JK, Ettinger A, Cruz-Pacheco N, Prochazka J, Haddox CL, Northrup E, Hodges C, Mostov KE, Hoffman MP, Knox SM. Parasympathetic innervation regulates tubulogenesis in the developing salivary gland. Dev Cell 2014; 30:449-62. [PMID: 25158854 DOI: 10.1016/j.devcel.2014.06.012] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 04/25/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022]
Abstract
A fundamental question in development is how cells assemble to form a tubular network during organ formation. In glandular organs, tubulogenesis is a multistep process requiring coordinated proliferation, polarization and reorganization of epithelial cells to form a lumen, and lumen expansion. Although it is clear that epithelial cells possess an intrinsic ability to organize into polarized structures, the mechanisms coordinating morphogenetic processes during tubulogenesis are poorly understood. Here, we demonstrate that parasympathetic nerves regulate tubulogenesis in the developing salivary gland. We show that vasoactive intestinal peptide (VIP) secreted by the innervating ganglia promotes ductal growth, leads to the formation of a contiguous lumen, and facilitates lumen expansion through a cyclic AMP/protein kinase A (cAMP/PKA)-dependent pathway. Furthermore, we provide evidence that lumen expansion is independent of apoptosis and involves the cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-regulated Cl(-) channel. Thus, parasympathetic innervation coordinates multiple steps in tubulogenesis during organogenesis.
Collapse
Affiliation(s)
- Pavel I Nedvetsky
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elaine Emmerson
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jennifer K Finley
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andreas Ettinger
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Noel Cruz-Pacheco
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jan Prochazka
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Candace L Haddox
- National Institute of Dental and Craniofacial Research, Bethesda, MD 20892, USA
| | - Emily Northrup
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Craig Hodges
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Keith E Mostov
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew P Hoffman
- National Institute of Dental and Craniofacial Research, Bethesda, MD 20892, USA
| | - Sarah M Knox
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
50
|
Woods E, Courtney J, Scholz D, Hall WW, Gautier VW. Tracking protein dynamics with photoconvertible Dendra2 on spinning disk confocal systems. J Microsc 2014; 256:197-207. [PMID: 25186063 DOI: 10.1111/jmi.12172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 07/31/2014] [Indexed: 01/25/2023]
Abstract
Understanding the dynamic properties of cellular proteins in live cells and in real time is essential to delineate their function. In this context, we introduce the Fluorescence Recovery After Photobleaching-Photoactivation unit (Andor) combined with the Nikon Eclipse Ti E Spinning Disk (Andor) confocal microscope as an advantageous and robust platform to exploit the properties of the Dendra2 photoconvertible fluorescent protein (Evrogen) and analyse protein subcellular trafficking in living cells. A major advantage of the spinning disk confocal is the rapid acquisition speed, enabling high temporal resolution of cellular processes. Furthermore, photoconversion and imaging are less invasive on the spinning disk confocal as the cell exposition to illumination power is reduced, thereby minimizing photobleaching and increasing cell viability. We have tested this commercially available platform using experimental settings adapted to track the migration of fast trafficking proteins such as UBC9, Fibrillarin and have successfully characterized their differential motion between subnuclear structures. We describe here step-by-step procedures, with emphasis on cellular imaging parameters, to successfully perform the dynamic imaging and photoconversion of Dendra2-fused proteins at high spatial and temporal resolutions necessary to characterize the trafficking pathways of proteins.
Collapse
Affiliation(s)
- Elena Woods
- Centre for Research in Infectious Diseases, School of Medicine and Biomedical Science, University College Dublin (UCD), Dublin, Ireland
| | | | | | | | | |
Collapse
|