101
|
Isogai T, Danuser G. Discovery of functional interactions among actin regulators by analysis of image fluctuations in an unperturbed motile cell system. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0110. [PMID: 29632262 DOI: 10.1098/rstb.2017.0110] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 01/06/2023] Open
Abstract
Cell migration is driven by propulsive forces derived from polymerizing actin that pushes and extends the plasma membrane. The underlying actin network is constantly undergoing adaptation to new mechano-chemical environments and intracellular conditions. As such, mechanisms that regulate actin dynamics inherently contain multiple feedback loops and redundant pathways. Given the highly adaptable nature of such a system, studies that use only perturbation experiments (e.g. knockdowns, overexpression, pharmacological activation/inhibition, etc.) are challenged by the nonlinearity and redundancy of the pathway. In these pathway configurations, perturbation experiments at best describe the function(s) of a molecular component in an adapting (e.g. acutely drug-treated) or fully adapted (e.g. permanent gene silenced) cell system, where the targeted component now resides in a non-native equilibrium. Here, we propose how quantitative live-cell imaging and analysis of constitutive fluctuations of molecular activities can overcome these limitations. We highlight emerging actin filament barbed-end biology as a prime example of a complex, nonlinear molecular process that requires a fluctuation analytic approach, especially in an unperturbed cellular system, to decipher functional interactions of barbed-end regulators, actin polymerization and membrane protrusion.This article is part of the theme issue 'Self-organization in cell biology'.
Collapse
Affiliation(s)
- Tadamoto Isogai
- Department of Cell Biology, Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gaudenz Danuser
- Department of Cell Biology, Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
102
|
Onwubiko UN, Mlynarczyk PJ, Wei B, Habiyaremye J, Clack A, Abel SM, Das ME. A Cdc42 GEF, Gef1, through endocytosis organizes F-BAR Cdc15 along the actomyosin ring and promotes concentric furrowing. J Cell Sci 2019; 132:jcs223776. [PMID: 30709916 PMCID: PMC6432710 DOI: 10.1242/jcs.223776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 01/21/2019] [Indexed: 01/17/2023] Open
Abstract
During cytokinesis, fission yeast coordinates actomyosin ring constriction with septum ingression, resulting in concentric furrow formation by a poorly defined mechanism. We report that Schizosaccharomyces pombe cells lacking the Cdc42 activator Gef1, combined with an activated allele of the formin, Cdc12, display non-concentric furrowing. Non-concentrically furrowing cells display uneven distribution of the scaffold Cdc15 along the ring. This suggests that, after ring assembly, uniform Cdc15 distribution along the ring enables proper furrow formation. We find that, after assembly, Cdc15 is recruited to the ring in an Arp2/3 complex-dependent manner and is decreased in the activated cdc12 mutant. Cdc15 at cortical endocytic patches shows increased levels and extended lifetimes in gef1 and activated cdc12 mutants. We hypothesize endocytosis helps recruit Cdc15 to assembled rings; uneven Cdc15 distribution at the ring occurs when endocytic patches contain increased Cdc15 levels and the patch-association rate is slow. Based on this, we developed a mathematical model that captures experimentally observed Cdc15 distributions along the ring. We propose that, at the ring, Gef1 and endocytic events promote uniform Cdc15 organization to enable proper septum ingression and concentric furrow formation.
Collapse
Affiliation(s)
- Udo N Onwubiko
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Paul J Mlynarczyk
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Bin Wei
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Julius Habiyaremye
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Amanda Clack
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Steven M Abel
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Maitreyi E Das
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
103
|
Fernández-Calleja V, Fernández-Nestosa MJ, Hernández P, Schvartzman JB, Krimer DB. CRISPR/Cas9-mediated deletion of the Wiskott-Aldrich syndrome locus causes actin cytoskeleton disorganization in murine erythroleukemia cells. PeerJ 2019; 7:e6284. [PMID: 30671311 PMCID: PMC6339507 DOI: 10.7717/peerj.6284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/14/2018] [Indexed: 01/18/2023] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is a recessive X-linked inmmunodeficiency caused by loss-of-function mutations in the gene encoding the WAS protein (WASp). WASp plays an important role in the polymerization of the actin cytoskeleton in hematopoietic cells through activation of the Arp2/3 complex. In a previous study, we found that actin cytoskeleton proteins, including WASp, were silenced in murine erythroleukemia cells defective in differentiation. Here, we designed a CRISPR/Cas9 strategy to delete a 9.5-kb genomic region encompassing the Was gene in the X chromosome of murine erythroleukemia (MEL) cells. We show that Was-deficient MEL cells have a poor organization of the actin cytoskeleton that can be recovered by restoring Was expression. We found that whereas the total amount of actin protein was similar between wild-type and Was knockout MEL cells, the latter exhibited an altered ratio of monomeric G-actin to polymeric F-actin. We also demonstrate that Was overexpression can mediate the activation of Bruton’s tyrosine kinase. Overall, these findings support the role of WASp as a key regulator of F-actin in erythroid cells.
Collapse
Affiliation(s)
- Vanessa Fernández-Calleja
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, Spanish National Research Council (CSIC), Madrid, Spain
| | | | - Pablo Hernández
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, Spanish National Research Council (CSIC), Madrid, Spain
| | - Jorge B Schvartzman
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, Spanish National Research Council (CSIC), Madrid, Spain
| | - Dora B Krimer
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
104
|
Shirakawa J, Kajikawa S, Böttcher RT, Costell M, Izu Y, Hayata T, Noda M, Ezura Y. Profilin 1 Negatively Regulates Osteoclast Migration in Postnatal Skeletal Growth, Remodeling, and Homeostasis in Mice. JBMR Plus 2019; 3:e10130. [PMID: 31346562 DOI: 10.1002/jbm4.10130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/16/2018] [Accepted: 10/21/2018] [Indexed: 01/29/2023] Open
Abstract
Profilin 1 (Pfn1), a regulator of actin polymerization, controls cell movement in a context-dependent manner. Pfn1 supports the locomotion of most adherent cells by assisting actin-filament elongation, as has been shown in skeletal progenitor cells in our previous study. However, because Pfn1 has also been known to inhibit migration of certain cells, including T cells, by suppressing branched-end elongation of actin filaments, we hypothesized that its roles in osteoclasts may be different from that of osteoblasts. By investigating the osteoclasts in culture, we first verified that Pfn1-knockdown (KD) enhances bone resorption in preosteoclastic RAW264.7 cells, despite having a comparable number and size of osteoclasts. Pfn1-KD in bone marrow cells showed similar results. Mechanistically, Pfn1-KD osteoclasts appeared more mobile than in controls. In vivo, the osteoclast-specific conditional Pfn1-deficient mice (Pfn1-cKO) by CathepsinK-Cre driver demonstrated postnatal skeletal phenotype, including dwarfism, craniofacial deformities, and long-bone metaphyseal osteolytic expansion, by 8 weeks of age. Metaphyseal and diaphyseal femurs were drastically expanded with suppressed trabecular bone mass as indicated by μCT analysis. Histologically, TRAP-positive osteoclasts were increased at endosteal metaphysis to diaphysis of Pfn1-cKO mice. The enhanced movement of Pfn1-cKO osteoclasts in culture was associated with a slight increase in cell size and podosome belt length, as well as an increase in bone-resorbing activity. Our study, for the first time, demonstrated that Pfn1 has critical roles in inhibiting osteoclast motility and bone resorption, thereby contributing to essential roles in postnatal skeletal homeostasis. Our study also provides novel insight into understanding skeletal deformities in human disorders.
Collapse
Affiliation(s)
- Jumpei Shirakawa
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Department of Oral Medicine and Stomatology School of Dental Medicine Tsurumi University Yokohama Japan
| | - Shuhei Kajikawa
- Frontier Research Unit Skeletal Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| | - Ralph T Böttcher
- Department of Molecular Medicine Max Planck Institute of Biochemistry Martinsried Germany
| | - Mercedes Costell
- Department of Biochemistry and Molecular Biology Faculty of Biology University of Valencia Spain
| | - Yayoi Izu
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| | - Tadayoshi Hayata
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Department of Molecular Pharmacology Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Science Tokyo University of Science Noda CHIBA Japan
| | - Masaki Noda
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Yokohama City Minato Red Cross Hospital Yokohama Japan.,Department of Orthopedic Surgery Tokyo Medical and Dental University Tokyo Japan
| | - Yoichi Ezura
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Frontier Research Unit Skeletal Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| |
Collapse
|
105
|
Pintard L, Bowerman B. Mitotic Cell Division in Caenorhabditis elegans. Genetics 2019; 211:35-73. [PMID: 30626640 PMCID: PMC6325691 DOI: 10.1534/genetics.118.301367] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/24/2018] [Indexed: 11/18/2022] Open
Abstract
Mitotic cell divisions increase cell number while faithfully distributing the replicated genome at each division. The Caenorhabditis elegans embryo is a powerful model for eukaryotic cell division. Nearly all of the genes that regulate cell division in C. elegans are conserved across metazoan species, including humans. The C. elegans pathways tend to be streamlined, facilitating dissection of the more redundant human pathways. Here, we summarize the virtues of C. elegans as a model system and review our current understanding of centriole duplication, the acquisition of pericentriolar material by centrioles to form centrosomes, the assembly of kinetochores and the mitotic spindle, chromosome segregation, and cytokinesis.
Collapse
Affiliation(s)
- Lionel Pintard
- Equipe labellisée Ligue contre le Cancer, Institut Jacques Monod, Team Cell Cycle and Development UMR7592, Centre National de la Recherche Scientifique - Université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France
| | - Bruce Bowerman
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| |
Collapse
|
106
|
Schell C, Sabass B, Helmstaedter M, Geist F, Abed A, Yasuda-Yamahara M, Sigle A, Maier JI, Grahammer F, Siegerist F, Artelt N, Endlich N, Kerjaschki D, Arnold HH, Dengjel J, Rogg M, Huber TB. ARP3 Controls the Podocyte Architecture at the Kidney Filtration Barrier. Dev Cell 2018; 47:741-757.e8. [PMID: 30503751 PMCID: PMC6302147 DOI: 10.1016/j.devcel.2018.11.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 09/03/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022]
Abstract
Podocytes, highly specialized epithelial cells, build the outer part of the kidney filtration barrier and withstand high mechanical forces through a complex network of cellular protrusions. Here, we show that Arp2/3-dependent actin polymerization controls actomyosin contractility and focal adhesion maturation of podocyte protrusions and thereby regulates formation, maintenance, and capacity to adapt to mechanical requirements of the filtration barrier. We find that N-WASP-Arp2/3 define the development of complex arborized podocyte protrusions in vitro and in vivo. Loss of dendritic actin networks results in a pronounced activation of the actomyosin cytoskeleton and the generation of over-maturated but less efficient adhesion, leading to detachment of podocytes. Our data provide a model to explain podocyte protrusion morphology and their mechanical stability based on a tripartite relationship between actin polymerization, contractility, and adhesion. ARP3-dependent actin assembly is required for podocyte process formation Arp2/3 thereby links process formation, podocyte adhesion and mechano-adaptation Arp2/3 function is regulated by a reciprocal interplay with actomyosin
Collapse
Affiliation(s)
- Christoph Schell
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Berta-Ottenstein Programme, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Benedikt Sabass
- Institute of Complex Systems-2, Forschungszentrum Jülich, Jülich 52428, Germany
| | - Martin Helmstaedter
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Felix Geist
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Ahmed Abed
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Mako Yasuda-Yamahara
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - August Sigle
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Jasmin I Maier
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Florian Grahammer
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald 17487, Germany
| | - Nadine Artelt
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald 17487, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald 17487, Germany
| | | | - Hans-Henning Arnold
- Cell and Molecular Biology, Technical University of Braunschweig, Braunschweig 38106, Germany
| | - Jörn Dengjel
- BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg 79106, Germany; Department of Biology, University of Fribourg, Fribourg 1700, Switzerland; Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Manuel Rogg
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Tobias B Huber
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg 79106, Germany.
| |
Collapse
|
107
|
Mullins RD, Bieling P, Fletcher DA. From solution to surface to filament: actin flux into branched networks. Biophys Rev 2018; 10:1537-1551. [PMID: 30470968 DOI: 10.1007/s12551-018-0469-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/21/2018] [Indexed: 02/05/2023] Open
Abstract
The actin cytoskeleton comprises a set of filament networks that perform essential functions in eukaryotic cells. The idea that actin filaments incorporate monomers directly from solution forms both the "textbook picture" of filament elongation and a conventional starting point for quantitative modeling of cellular actin dynamics. Recent work, however, reveals that filaments created by two major regulators, the formins and the Arp2/3 complex, incorporate monomers delivered by nearby proteins. Specifically, actin enters Arp2/3-generated networks via binding sites on nucleation-promoting factors clustered on membrane surfaces. Here, we describe three functions of this surface-associated actin monomer pool: (1) regulating network density via product inhibition of the Arp2/3 complex, (2) accelerating filament elongation as a distributive polymerase, and (3) converting profilin-actin into a substrate for the Arp2/3 complex. These linked functions control the architecture of branched networks and explain how capping protein enhances their growth.
Collapse
Affiliation(s)
- R Dyche Mullins
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, UCSF School of Medicine, San Francisco, CA, USA.
| | - Peter Bieling
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Daniel A Fletcher
- Department of Bioengineering, University of California, Berkeley, CA, USA
| |
Collapse
|
108
|
Chan FY, Silva AM, Saramago J, Pereira-Sousa J, Brighton HE, Pereira M, Oegema K, Gassmann R, Carvalho AX. The ARP2/3 complex prevents excessive formin activity during cytokinesis. Mol Biol Cell 2018; 30:96-107. [PMID: 30403552 PMCID: PMC6337913 DOI: 10.1091/mbc.e18-07-0471] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cytokinesis completes cell division by constriction of an actomyosin contractile ring that separates the two daughter cells. Here we use the early Caenorhabditis elegans embryo to explore how the actin filament network in the ring and the surrounding cortex is regulated by the single cytokinesis formin CYK-1 and the ARP2/3 complex, which nucleate nonbranched and branched filaments, respectively. We show that CYK-1 and the ARP2/3 complex are the predominant F-actin nucleators responsible for generating distinct cortical F-actin architectures and that depletion of either nucleator affects the kinetics of cytokinesis. CYK-1 is critical for normal F-actin levels in the contractile ring, and acute inhibition of CYK-1 after furrow ingression slows ring constriction rate, suggesting that CYK-1 activity is required throughout ring constriction. Surprisingly, although the ARP2/3 complex does not localize in the contractile ring, depletion of the ARP2 subunit or treatment with ARP2/3 complex inhibitor delays contractile ring formation and constriction. We present evidence that the delays are due to an excess in formin-nucleated cortical F-actin, suggesting that the ARP2/3 complex negatively regulates CYK-1 activity. We conclude that the kinetics of cytokinesis are modulated by interplay between the two major actin filament nucleators.
Collapse
Affiliation(s)
- Fung-Yi Chan
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana M Silva
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Saramago
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Pereira-Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Hailey E Brighton
- Department of Cellular and Molecular Medicine, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093
| | - Marisa Pereira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Karen Oegema
- Department of Cellular and Molecular Medicine, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093
| | - Reto Gassmann
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Xavier Carvalho
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.,Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
109
|
Courtemanche N. Mechanisms of formin-mediated actin assembly and dynamics. Biophys Rev 2018; 10:1553-1569. [PMID: 30392063 DOI: 10.1007/s12551-018-0468-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
Cellular viability requires tight regulation of actin cytoskeletal dynamics. Distinct families of nucleation-promoting factors enable the rapid assembly of filament nuclei that elongate and are incorporated into diverse and specialized actin-based structures. In addition to promoting filament nucleation, the formin family of proteins directs the elongation of unbranched actin filaments. Processive association of formins with growing filament ends is achieved through continuous barbed end binding of the highly conserved, dimeric formin homology (FH) 2 domain. In cooperation with the FH1 domain and C-terminal tail region, FH2 dimers mediate actin subunit addition at speeds that can dramatically exceed the rate of spontaneous assembly. Here, I review recent biophysical, structural, and computational studies that have provided insight into the mechanisms of formin-mediated actin assembly and dynamics.
Collapse
Affiliation(s)
- Naomi Courtemanche
- Department of Genetics, Cell and Developmental Biology, University of Minnesota, 420 Washington Ave SE, 6-130 MCB, Minneapolis, MN, 55455, USA.
| |
Collapse
|
110
|
Assembling actin filaments for protrusion. Curr Opin Cell Biol 2018; 56:53-63. [PMID: 30278304 DOI: 10.1016/j.ceb.2018.09.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/31/2022]
Abstract
Cell migration entails a plethora of activities combining the productive exertion of protrusive and contractile forces to allow cells to push and squeeze themselves through cell clumps, interstitial tissues or tissue borders. All these activities require the generation and turnover of actin filaments that arrange into specific, subcellular structures. The most prominent structures mediating the protrusion at the leading edges of cells include lamellipodia and filopodia as well as plasma membrane blebs. Moreover, in cells migrating on planar substratum, mechanical support is being provided by an additional, more proximally located structure termed the lamella. Here, we systematically dissect the literature concerning the mechanisms driving actin filament nucleation and elongation in the best-studied protrusive structure, the lamellipodium. Recent work has shed light on open questions in lamellipodium protrusion, including the relative contributions of nucleation versus elongation to the assembly of both individual filaments and the lamellipodial network as a whole. However, much remains to be learned concerning the specificity and relevance of individual factors, their cooperation and their site-specific functions relative to the importance of global actin monomer and filament homeostasis.
Collapse
|
111
|
Roy NH, Burkhardt JK. The Actin Cytoskeleton: A Mechanical Intermediate for Signal Integration at the Immunological Synapse. Front Cell Dev Biol 2018; 6:116. [PMID: 30283780 PMCID: PMC6156151 DOI: 10.3389/fcell.2018.00116] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/27/2018] [Indexed: 12/22/2022] Open
Abstract
The immunological synapse (IS) is a specialized structure that serves as a platform for cell-cell communication between a T cell and an antigen-presenting cell (APC). Engagement of the T cell receptor (TCR) with cognate peptide-MHC complexes on the APC activates the T cell and instructs its differentiation. Proper T cell activation also requires engagement of additional receptor-ligand pairs, which promote sustained adhesion and deliver costimulatory signals. These events are orchestrated by T cell actin dynamics, which organize IS components and facilitate their signaling. The actin network flows from the edge of the cell inward, driving the centralization of TCR microclusters and providing the force to activate the integrin LFA-1. We recently showed that engagement of LFA-1 slows actin flow, and that this affects TCR signaling. This study highlights the physical nature of the IS, and contributes to a growing appreciation in the field that mechanosensing and mechanotransduction are essential for IS function. Additionally, it is becoming clear that there are multiple types of actin structures at the IS that promote signaling in distinct ways. How the different actin structures contribute to force production and mechanotransduction is just beginning to be explored. In this Perspective, we will feature recent work from our lab and others, that collectively points toward a model in which actin dynamics drive mechanical signaling and receptor crosstalk during T cell activation.
Collapse
Affiliation(s)
- Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
112
|
Plastino J, Blanchoin L. Dynamic stability of the actin ecosystem. J Cell Sci 2018; 132:132/4/jcs219832. [PMID: 30104258 DOI: 10.1242/jcs.219832] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
In cells, actin filaments continuously assemble and disassemble while maintaining an apparently constant network structure. This suggests a perfect balance between dynamic processes. Such behavior, operating far out of equilibrium by the hydrolysis of ATP, is called a dynamic steady state. This dynamic steady state confers a high degree of plasticity to cytoskeleton networks that allows them to adapt and optimize their architecture in response to external changes on short time-scales, thus permitting cells to adjust to their environment. In this Review, we summarize what is known about the cellular actin steady state, and what gaps remain in our understanding of this fundamental dynamic process that balances the different forms of actin organization in a cell. We focus on the minimal steps to achieve a steady state, discuss the potential feedback mechanisms at play to balance this steady state and conclude with an outlook on what is needed to fully understand its molecular nature.
Collapse
Affiliation(s)
- Julie Plastino
- Institut Curie, PSL Research University, CNRS, 75005 Paris, France .,Sorbonne Université, 75005 Paris, France
| | - Laurent Blanchoin
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054 Grenoble, France .,CytomorphoLab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, UMRS1160, INSERM/AP-HP/Université Paris Diderot, 75010 Paris, France
| |
Collapse
|
113
|
Kawabata Galbraith K, Fujishima K, Mizuno H, Lee SJ, Uemura T, Sakimura K, Mishina M, Watanabe N, Kengaku M. MTSS1 Regulation of Actin-Nucleating Formin DAAM1 in Dendritic Filopodia Determines Final Dendritic Configuration of Purkinje Cells. Cell Rep 2018; 24:95-106.e9. [DOI: 10.1016/j.celrep.2018.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/01/2018] [Accepted: 06/01/2018] [Indexed: 10/28/2022] Open
|
114
|
Martín-Cófreces NB, Sánchez-Madrid F. Sailing to and Docking at the Immune Synapse: Role of Tubulin Dynamics and Molecular Motors. Front Immunol 2018; 9:1174. [PMID: 29910809 PMCID: PMC5992405 DOI: 10.3389/fimmu.2018.01174] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 12/17/2022] Open
Abstract
The different cytoskeleton systems and their connecting molecular motors move vesicles and intracellular organelles to shape cells. Polarized cells with specialized functions display an exquisite spatio-temporal regulation of both cytoskeletal and organelle arrangements that support their specific tasks. In particular, T cells rapidly change their shape and cellular function through the establishment of cell surface and intracellular polarity in response to a variety of cues. This review focuses on the contribution of the microtubule-based dynein/dynactin motor complex, the tubulin and actin cytoskeletons, and different organelles to the formation of the antigen-driven immune synapse.
Collapse
Affiliation(s)
- Noa Beatriz Martín-Cófreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
115
|
Kotila T, Kogan K, Enkavi G, Guo S, Vattulainen I, Goode BL, Lappalainen P. Structural basis of actin monomer re-charging by cyclase-associated protein. Nat Commun 2018; 9:1892. [PMID: 29760438 PMCID: PMC5951797 DOI: 10.1038/s41467-018-04231-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/13/2018] [Indexed: 11/10/2022] Open
Abstract
Actin polymerization powers key cellular processes, including motility, morphogenesis, and endocytosis. The actin turnover cycle depends critically on "re-charging" of ADP-actin monomers with ATP, but whether this reaction requires dedicated proteins in cells, and the underlying mechanism, have remained elusive. Here we report that nucleotide exchange catalyzed by the ubiquitous cytoskeletal regulator cyclase-associated protein (CAP) is critical for actin-based processes in vivo. We determine the structure of the CAP-actin complex, which reveals that nucleotide exchange occurs in a compact, sandwich-like complex formed between the dimeric actin-binding domain of CAP and two ADP-actin monomers. In the crystal structure, the C-terminal tail of CAP associates with the nucleotide-sensing region of actin, and this interaction is required for rapid re-charging of actin by both yeast and mammalian CAPs. These data uncover the conserved structural basis and biological role of protein-catalyzed re-charging of actin monomers.
Collapse
Affiliation(s)
- Tommi Kotila
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Konstantin Kogan
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Giray Enkavi
- Department of Physics, University of Helsinki, 00014, Helsinki, Finland
| | - Siyang Guo
- Department of Biology, Brandeis University, Waltham, MA, 02453, USA
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, 00014, Helsinki, Finland
- Laboratory of Physics, Tampere University of Technology, 33101, Tampere, Finland
| | - Bruce L Goode
- Department of Biology, Brandeis University, Waltham, MA, 02453, USA
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
116
|
Segal D, Zaritsky A, Schejter ED, Shilo BZ. Feedback inhibition of actin on Rho mediates content release from large secretory vesicles. J Cell Biol 2018; 217:1815-1826. [PMID: 29496739 PMCID: PMC5940311 DOI: 10.1083/jcb.201711006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/24/2017] [Accepted: 01/30/2018] [Indexed: 12/02/2022] Open
Abstract
Secretion of adhesive glycoproteins to the lumen of Drosophila melanogaster larval salivary glands is performed by contraction of an actomyosin network assembled around large secretory vesicles, after their fusion to the apical membranes. We have identified a cycle of actin coat nucleation and disassembly that is independent of myosin. Recruitment of active Rho1 to the fused vesicle triggers activation of the formin Diaphanous and actin nucleation. This leads to actin-dependent localization of a RhoGAP protein that locally shuts off Rho1, promoting disassembly of the actin coat. When contraction of vesicles is blocked, the strict temporal order of the recruited elements generates repeated oscillations of actin coat formation and disassembly. Interestingly, different blocks to actin coat disassembly arrested vesicle contraction, indicating that actin turnover is an integral part of the actomyosin contraction cycle. The capacity of F-actin to trigger a negative feedback on its own production may be widely used to coordinate a succession of morphogenetic events or maintain homeostasis.
Collapse
Affiliation(s)
- Dagan Segal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Assaf Zaritsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
117
|
Actin Cross-Linking Toxin Is a Universal Inhibitor of Tandem-Organized and Oligomeric G-Actin Binding Proteins. Curr Biol 2018; 28:1536-1547.e9. [PMID: 29731300 DOI: 10.1016/j.cub.2018.03.065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/06/2018] [Accepted: 03/28/2018] [Indexed: 11/20/2022]
Abstract
Delivery of bacterial toxins to host cells is hindered by host protective barriers. This obstruction dictates a remarkable efficiency of toxins, a single copy of which may kill a host cell. Efficiency of actin-targeting toxins is further hampered by an overwhelming abundance of their target. The actin cross-linking domain (ACD) toxins of Vibrio species and related bacterial genera catalyze the formation of covalently cross-linked actin oligomers. Recently, we reported that the ACD toxicity can be amplified via a multivalent inhibitory association of actin oligomers with actin assembly factors formins, suggesting that the oligomers may act as secondary toxins. Importantly, many proteins involved in nucleation, elongation, severing, branching, and bundling of actin filaments contain G-actin-binding Wiskott-Aldrich syndrome protein (WASP)-homology motifs 2 (WH2) organized in tandem and therefore may act as a multivalent platform for high-affinity interaction with the ACD-cross-linked actin oligomers. Using live-cell single-molecule speckle (SiMS) microscopy, total internal reflection fluorescence (TIRF) microscopy, and actin polymerization assays, we show that, in addition to formins, the oligomers bind with high affinity and potently inhibit several families of actin assembly factors: Ena/vasodilator-stimulated phosphorprotein (VASP); Spire; and the Arp2/3 complex, both in vitro and in live cells. As a result, ACD blocks the actin retrograde flow and membrane dynamics and disrupts association of Ena/VASP with adhesion complexes. This study defines ACD as a universal inhibitor of tandem-organized G-actin binding proteins that overcomes the abundance of actin by redirecting the toxicity cascade toward less abundant targets and thus leading to profound disorganization of the actin cytoskeleton and disruption of actin-dependent cellular functions.
Collapse
|
118
|
Zhang MS, Tran PM, Wolff AJ, Tremblay MM, Fosdick MG, Houtman JCD. Glycerol monolaurate induces filopodia formation by disrupting the association between LAT and SLP-76 microclusters. Sci Signal 2018; 11:11/528/eaam9095. [PMID: 29717064 DOI: 10.1126/scisignal.aam9095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glycerol monolaurate (GML) is a monoglyceride with potent antimicrobial properties that suppresses T cell receptor (TCR)-induced signaling and T cell effector function. Actin rearrangement is needed for the interaction of T cells with antigen-presenting cells and for migration to sites of infection. Because of the critical role actin rearrangement plays in T cell effector function, we analyzed the effect of GML on the rearrangement of the actin cytoskeleton after TCR activation. We found that GML-treated human T cells were less adherent than untreated T cells and did not form actin ring structures but instead developed numerous inappropriate actin-mediated filopodia. The formation of these filopodia was not due to disruption of TCR-proximal regulators of actin or microtubule polymerization. Instead, total internal reflection fluorescence microscopy demonstrated mislocalization of actin nucleation protein Arp2 microclusters, but not those containing the adaptor proteins SLP-76 and WASp, or the actin nucleation protein ARPC3, which are necessary for TCR-induced actin rearrangement. Additionally, SLP-76 microclusters colocalized with WASp and WAVE microclusters but not with LAT. Together, our data suggest that GML alters actin cytoskeletal rearrangements and identify diverse functions for GML as a T cell-suppressive agent.
Collapse
Affiliation(s)
- Michael S Zhang
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Phuong M Tran
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Alexander J Wolff
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Mikaela M Tremblay
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Micaela G Fosdick
- Biomedical Sciences Program, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jon C D Houtman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA. .,Biomedical Sciences Program, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
119
|
Capping protein-controlled actin polymerization shapes lipid membranes. Nat Commun 2018; 9:1630. [PMID: 29691404 PMCID: PMC5915599 DOI: 10.1038/s41467-018-03918-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 03/20/2018] [Indexed: 11/08/2022] Open
Abstract
Arp2/3 complex-mediated actin assembly at cell membranes drives the formation of protrusions or endocytic vesicles. To identify the mechanism by which different membrane deformations can be achieved, we reconstitute the basic membrane deformation modes of inward and outward bending in a confined geometry by encapsulating a minimal set of cytoskeletal proteins into giant unilamellar vesicles. Formation of membrane protrusions is favoured at low capping protein (CP) concentrations, whereas the formation of negatively bent domains is promoted at high CP concentrations. Addition of non-muscle myosin II results in full fission events in the vesicle system. The different deformation modes are rationalized by simulations of the underlying transient nature of the reaction kinetics. The relevance of the regulatory mechanism is supported by CP overexpression in mouse melanoma B16-F1 cells and therefore demonstrates the importance of the quantitative understanding of microscopic kinetic balances to address the diverse functionality of the cytoskeleton. Cell membrane protrusions and invaginations are both driven by actin assembly but the mechanism leading to different membrane shapes is unknown. Using a minimal system and modelling the authors reconstitute the deformation modes and identify capping protein as a regulator of both deformation types.
Collapse
|
120
|
Fu Y, Yu W, Cai H, Lu A. Forecast of actin-binding proteins as the oncotarget in osteosarcoma - a review of mechanism, diagnosis and therapy. Onco Targets Ther 2018; 11:1553-1561. [PMID: 29593421 PMCID: PMC5865567 DOI: 10.2147/ott.s159894] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone malignant tumor with a high rate of lung metastasis and principally emerges in children and adolescents. Although neoadjuvant chemotherapy is widely used around the world, a high rate of chemoresistance occurs and frequently generates a poor prognosis. Therefore, finding a new appropriate prognostic marker for OS is a valuable research direction, which will give patients a better chance to receive proper therapy. Actin-binding proteins (ABPs) are a group of proteins that interact with actin cytoskeleton and play a crucial role in the regulation of the cell motility and morphology in eukaryotes. Meanwhile, ABPs also act as a bridge between the cytomembrane and nucleus, which transmit the outside-in and inside-out signals in cytoplasm. Furthermore, ABPs alter the dynamic structure of actin and regulate the invasion and metastasis of cancer. Hence, ABPs have a wide application in predicting the prognosis, and may be new targets, in tumor therapy. This review focuses on a series of ABPs and discusses their modulatory functions. It provides a new insight into the classification of ABPs’ functions in the process of invasion and metastasis in OS and illuminates the potential ability in predicting the prognosis of OS patients.
Collapse
Affiliation(s)
- Yucheng Fu
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Wei Yu
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Hongliu Cai
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Anwei Lu
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
121
|
Leelarasamee N, Zhang L, Gleason C. The root-knot nematode effector MiPFN3 disrupts plant actin filaments and promotes parasitism. PLoS Pathog 2018; 14:e1006947. [PMID: 29543900 PMCID: PMC5871015 DOI: 10.1371/journal.ppat.1006947] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/27/2018] [Accepted: 02/21/2018] [Indexed: 12/03/2022] Open
Abstract
Root-knot nematodes secrete effectors that manipulate their host plant cells so that the nematode can successfully establish feeding sites and complete its lifecycle. The root-knot nematode feeding structures, their “giant cells,” undergo extensive cytoskeletal remodeling. Previous cytological studies have shown the cytoplasmic actin within the feeding sites looks diffuse. In an effort to study root-knot nematode effectors that are involved in giant cell organogenesis, we have identified a nematode effector called MiPFN3 (Meloidogyne incognita Profilin 3). MiPFN3 is transcriptionally up-regulated in the juvenile stage of the nematode. In situ hybridization experiments showed that MiPFN3 transcribed in the nematode subventral glands, where it can be secreted by the nematode stylet into the plant. Moreover, Arabidopsis plants that heterologously expressed MiPFN3 were more susceptible to root-knot nematodes, indicating that MiPFN3 promotes nematode parasitism. Since profilin proteins can bind and sequester actin monomers, we investigated the function of MiPFN3 in relation to actin. Our results show that MiPFN3 suppressed the aberrant plant growth phenotype caused by the misexpression of reproductive actin (AtACT1) in transgenic plants. In addition, it disrupted actin polymerization in an in vitro assay, and it reduced the filamentous actin network when expressed in Arabidopsis protoplasts. Over a decade ago, cytological studies showed that the cytoplasmic actin within nematode giant cells looked fragmented. Here we provide the first evidence that the nematode is secreting an effector that has significant, direct effects on the plant’s actin cytoskeleton. Root-knot nematodes are microscopic plant pests that infect plant roots and significantly reduce yields of many crop plants. The nematodes enter the plant roots and modify plant cells into complex, multinuclear feeding sites called giant cells. The formation and maintenance of giant cells is critical to nematode survival. During giant cell organogenesis, the progenitor plant cells undergo many morphological changes, including a re-organization of the cytoplasmic actin cytoskeleton. As a result, the giant cell cytoplasmic actin appears fragmented and disorganized. Plant cells can regulate their actin filament assembly, in part, through the expression of actin binding proteins such as profilins. Here we show that infectious nematode juveniles express a profilin called MiPFN3. Expression of MiPFN3 in Arabidopsis plants made the plants more susceptible to root-knot nematodes, indicating that MiPFN3 acts as an effector that aids parasitism. We show evidence that the expression MiPFN3 in plant cells causes the fragmentation of plant actin filaments. The work here demonstrates that nematode effector MiPFN3 can directly affect plant actin filaments, whose reorganization is necessary for giant cell formation.
Collapse
Affiliation(s)
- Natthanon Leelarasamee
- Department of Plant Molecular Biology and Physiology, Albrecht von Haller Institute, Georg August University, Göttingen, Germany
| | - Lei Zhang
- Department of Plant Pathology, Washington State University, Pullman, WA, United States of America
| | - Cynthia Gleason
- Department of Plant Molecular Biology and Physiology, Albrecht von Haller Institute, Georg August University, Göttingen, Germany
- Department of Plant Pathology, Washington State University, Pullman, WA, United States of America
- * E-mail:
| |
Collapse
|
122
|
Abstract
Nitric oxide (NO) is a key messenger in the pathogenesis of inflammation, linking innate and adaptive immunity. By targeting signaling molecules, NO from inducible NO synthase (iNOS) and endothelial (e)NOS affects T helper cell differentiation and the effector functions of T lymphocytes, and is a potential target for therapeutic manipulation. In this review we discuss the regulatory actions exerted by NO on T cell functions, focusing on S-nitrosylation as an important post-translational modification by which NO acts as a signaling molecule during T cell-mediated immunity. We also present recent findings showing novel mechanisms through which NO regulates the activation of human T cells, and consider their potential in strategies to treat tumoral, allergic, and autoimmune diseases.
Collapse
|
123
|
Liu J, Wu Y, Wang Q, Liu X, Liao X, Pan J. Bioinformatic analysis of PFN2 dysregulation and its prognostic value in head and neck squamous carcinoma. Future Oncol 2018; 14:449-459. [PMID: 29322815 DOI: 10.2217/fon-2017-0348] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIM This study aimed to identify PFN2 expression profile, its prognostic value and the mechanism of its dysregulation in head and neck squamous cell carcinoma (HNSC). MATERIALS & METHODS Bioinformatic analysis was performed using data in the Gene Expression Omnibus Datasets, Human Protein Atlas and The Cancer Genome Atlas-HNSC. RESULTS PFN2 was upregulated in HNSC than in normal head and neck tissues. High PFN2 expression independently predicted poor overall survival in primary HNSC (hazard ratio: 1.548, 95% CI: 1.174-2.042; p = 0.002). Fourteen percent of HNSC cases had PFN2 amplification. PFN2 DNA methylation was negatively correlated with its mRNA expression (Pearson's r = -0.713). CONCLUSION High PFN2 expression might serve as a valuable predictor for poor overall survival of HNSC. DNA amplification and hypomethylation might be two mechanisms of PFN2 dysregulation.
Collapse
Affiliation(s)
- Jiyuan Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Clinical Research, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunlong Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Clinical Research, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qizhang Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Clinical Research, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Clinical Research, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xuejuan Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Clinical Research, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Clinical Research, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
124
|
Skruber K, Read TA, Vitriol EA. Reconsidering an active role for G-actin in cytoskeletal regulation. J Cell Sci 2018; 131:131/1/jcs203760. [PMID: 29321224 DOI: 10.1242/jcs.203760] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Globular (G)-actin, the actin monomer, assembles into polarized filaments that form networks that can provide structural support, generate force and organize the cell. Many of these structures are highly dynamic and to maintain them, the cell relies on a large reserve of monomers. Classically, the G-actin pool has been thought of as homogenous. However, recent work has shown that actin monomers can exist in distinct groups that can be targeted to specific networks, where they drive and modify filament assembly in ways that can have profound effects on cellular behavior. This Review focuses on the potential factors that could create functionally distinct pools of actin monomers in the cell, including differences between the actin isoforms and the regulation of G-actin by monomer binding proteins, such as profilin and thymosin β4. Owing to difficulties in studying and visualizing G-actin, our knowledge over the precise role that specific actin monomer pools play in regulating cellular actin dynamics remains incomplete. Here, we discuss some of these unanswered questions and also provide a summary of the methodologies currently available for the imaging of G-actin.
Collapse
Affiliation(s)
- Kristen Skruber
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Tracy-Ann Read
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Eric A Vitriol
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
125
|
Bascom CS, Hepler PK, Bezanilla M. Interplay between Ions, the Cytoskeleton, and Cell Wall Properties during Tip Growth. PLANT PHYSIOLOGY 2018; 176:28-40. [PMID: 29138353 PMCID: PMC5761822 DOI: 10.1104/pp.17.01466] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/05/2017] [Indexed: 05/08/2023]
Abstract
Tip growth is a focused and tightly regulated apical explosion that depends on the interconnected activities of ions, the cytoskeleton, and the cell wall.
Collapse
Affiliation(s)
- Carlisle S Bascom
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
- Plant Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01002
| | - Peter K Hepler
- Biology Department, University of Massachusetts, Amherst, Massachusetts 01002
| | - Magdalena Bezanilla
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| |
Collapse
|
126
|
PFN2a, a new partner of RARα in the cytoplasm. Biochem Biophys Res Commun 2018; 495:846-853. [PMID: 29158086 DOI: 10.1016/j.bbrc.2017.11.096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/15/2017] [Indexed: 11/23/2022]
Abstract
Retinoic acid receptors (RARs) are classically considered as nuclear ligand-dependent regulators of transcription. Here we highlighted a novel face of the RARα subtype: RARα is present in low amounts in the cytoplasm of mouse embryonic fibroblasts (MEFs) where it interacts with profilin2a (PFN2A), a small actin-binding protein involved in filaments polymerization. The interaction involves the N-terminal proline-rich motif (PRM) of RARα and the SH3-like domain of PFN2a. When increased in the cytoplasm, RARα competes with other PFN2a-binding proteins bearing PRMs and involved in actin filaments elongation. Consequently, the actin filament network is altered and MEFs adhesion is decreased. This novel role opens novel avenues for the understanding of pathologies characterized by increased levels of cytoplasmic RARα.
Collapse
|
127
|
Henty-Ridilla JL, Juanes MA, Goode BL. Profilin Directly Promotes Microtubule Growth through Residues Mutated in Amyotrophic Lateral Sclerosis. Curr Biol 2017; 27:3535-3543.e4. [PMID: 29129529 PMCID: PMC5772683 DOI: 10.1016/j.cub.2017.10.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/31/2017] [Accepted: 10/02/2017] [Indexed: 12/11/2022]
Abstract
Profilin is an abundant actin monomer-binding protein with critical actin regulatory roles in vivo [1, 2]. However, profilin also influences microtubule dynamics in cells, which may be mediated in part through its interactions with formins that in turn bind microtubules [3, 4]. Specific residues on human profilin-1 (PFN1) are mutated in patients with amyotrophic lateral sclerosis (ALS) [5, 6]. However, the observation that some ALS-linked PFN1 mutants fail to alter cellular actin organization or dynamics [5-8] or in vitro actin-monomer affinity [9] has been perplexing, given that profilin is best understood as an actin regulator. Here, we investigated direct effects of profilin on microtubule dynamics and whether ALS-linked mutations in PFN1 disrupt such functions. We found that human, fly, and yeast profilin homologs all directly enhance microtubule growth rate by several-fold in vitro. Microtubule stimulatory effects were unaffected by mutations in the canonical actin- or poly-proline-binding sites of profilin. Instead, microtubule activities depended on specific surface residues on profilin mutated in ALS patients. Furthermore, microtubule effects were attenuated by increasing concentrations of actin monomers, suggesting competition between actin and microtubules for binding profilin. Consistent with these biochemical observations, a 2-fold increase in the expression level of wild-type PFN1, but not the ALS-linked PFN1 mutants, increased microtubule growth rates in cells. Together, these results demonstrate that profilin directly enhances the growth rate of microtubules. They further suggest that ALS-linked mutations in PFN1 may perturb cellular microtubule dynamics and/or the coordination between the actin and microtubule cytoskeletons, leading to motor neuron degeneration.
Collapse
Affiliation(s)
| | - M Angeles Juanes
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Bruce L Goode
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| |
Collapse
|
128
|
Bieling P, Hansen SD, Akin O, Li TD, Hayden CC, Fletcher DA, Mullins RD. WH2 and proline-rich domains of WASP-family proteins collaborate to accelerate actin filament elongation. EMBO J 2017; 37:102-121. [PMID: 29141912 PMCID: PMC5753033 DOI: 10.15252/embj.201797039] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 02/04/2023] Open
Abstract
WASP‐family proteins are known to promote assembly of branched actin networks by stimulating the filament‐nucleating activity of the Arp2/3 complex. Here, we show that WASP‐family proteins also function as polymerases that accelerate elongation of uncapped actin filaments. When clustered on a surface, WASP‐family proteins can drive branched actin networks to grow much faster than they could by direct incorporation of soluble monomers. This polymerase activity arises from the coordinated action of two regulatory sequences: (i) a WASP homology 2 (WH2) domain that binds actin, and (ii) a proline‐rich sequence that binds profilin–actin complexes. In the absence of profilin, WH2 domains are sufficient to accelerate filament elongation, but in the presence of profilin, proline‐rich sequences are required to support polymerase activity by (i) bringing polymerization‐competent actin monomers in proximity to growing filament ends, and (ii) promoting shuttling of actin monomers from profilin–actin complexes onto nearby WH2 domains. Unoccupied WH2 domains transiently associate with free filament ends, preventing their growth and dynamically tethering the branched actin network to the WASP‐family proteins that create it. Collaboration between WH2 and proline‐rich sequences thus strikes a balance between filament growth and tethering. Our work expands the number of critical roles that WASP‐family proteins play in the assembly of branched actin networks to at least three: (i) promoting dendritic nucleation; (ii) linking actin networks to membranes; and (iii) accelerating filament elongation.
Collapse
Affiliation(s)
- Peter Bieling
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA .,Department of Bioengineering & Biophysics Program, University of California, Berkeley, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Scott D Hansen
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Orkun Akin
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Tai-De Li
- Department of Bioengineering & Biophysics Program, University of California, Berkeley, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA.,Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | - Daniel A Fletcher
- Department of Bioengineering & Biophysics Program, University of California, Berkeley, CA, USA .,Chan Zuckerberg Biohub, San Francisco, CA, USA.,Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
129
|
Proteomic features of delayed ocular symptoms caused by exposure to sulfur mustard: As studied by protein profiling of corneal epithelium. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1445-1454. [DOI: 10.1016/j.bbapap.2017.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022]
|
130
|
iTRAQ-Based Identification of Proteins Related to Muscle Growth in the Pacific Abalone, Haliotis discus hannai. Int J Mol Sci 2017; 18:ijms18112237. [PMID: 29068414 PMCID: PMC5713207 DOI: 10.3390/ijms18112237] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/20/2017] [Accepted: 10/21/2017] [Indexed: 11/16/2022] Open
Abstract
The abalone Haliotis discus hannai is an important aquaculture species that is grown for human consumption. However, little is known of the genetic mechanisms governing muscle growth in this species, particularly with respect to proteomics. The isobaric tag for relative and absolute quantitation (iTRAQ) method allows for sensitive and accurate protein quantification. Our study was the first to use iTRAQ-based quantitative proteomics to investigate muscle growth regulation in H. discus hannai. Among the 1904 proteins identified from six samples, 125 proteins were differentially expressed in large specimens of H. discus hannai as compared to small specimens. In the large specimens, 47 proteins were upregulated and 78 were downregulated. Many of the significant Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including these differentially expressed proteins, were closely related to muscle growth, including apoptosis, thyroid hormone signaling, regulation of the actin cytoskeleton, and viral myocarditis (p < 0.05). Our quantitative real-time polymerase chain reaction (qRT-PCR) analyses suggested that the alterations in expression levels observed in the differentially expressed proteins were consistent with the alterations observed in the encoding mRNAs, indicating the repeatability of our proteomic approach. Our findings contribute to the knowledge of the molecular mechanisms of muscle growth in H. discus hannai.
Collapse
|
131
|
14-3-3 Regulates Actin Filament Formation in the Deep-Branching Eukaryote Giardia lamblia. mSphere 2017; 2:mSphere00248-17. [PMID: 28932813 PMCID: PMC5597967 DOI: 10.1128/msphere.00248-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/21/2017] [Indexed: 01/30/2023] Open
Abstract
The phosphoserine/phosphothreonine-binding protein 14-3-3 is known to regulate actin; this function has been previously attributed to sequestration of phosphorylated cofilin. 14-3-3 was identified as an actin-associated protein in the deep-branching eukaryote Giardia lamblia; however, Giardia lacks cofilin and all other canonical actin-binding proteins (ABPs). Thus, the role of G. lamblia 14-3-3 (Gl-14-3-3) in actin regulation was unknown. Gl-14-3-3 depletion resulted in an overall disruption of actin organization characterized by ectopically distributed short actin filaments. Using phosphatase and kinase inhibitors, we demonstrated that actin phosphorylation correlated with destabilization of the actin network and increased complex formation with 14-3-3, while blocking actin phosphorylation stabilized actin filaments and attenuated complex formation. Giardia's sole Rho family GTPase, Gl-Rac, modulates Gl-14-3-3's association with actin, providing the first connection between Gl-Rac and the actin cytoskeleton in Giardia. Giardia actin (Gl-actin) contains two putative 14-3-3 binding motifs, one of which (S330) is conserved in mammalian actin. Mutation of these sites reduced, but did not completely disrupt, the association with 14-3-3. Native gels and overlay assays indicate that intermediate proteins are required to support complex formation between 14-3-3 and actin. Overall, our results support a role for 14-3-3 as a regulator of actin; however, the presence of multiple 14-3-3-actin complexes suggests a more complex regulatory relationship than might be expected for a minimalistic parasite. IMPORTANCEGiardia lacks canonical actin-binding proteins. Gl-14-3-3 was identified as an actin interactor, but the significance of this interaction was unknown. Loss of Gl-14-3-3 results in ectopic short actin filaments, indicating that Gl-14-3-3 is an important regulator of the actin cytoskeleton in Giardia. Drug studies indicate that Gl-14-3-3 complex formation is in part phospho-regulated. We demonstrate that complex formation is downstream of Giardia's sole Rho family GTPase, Gl-Rac. This result provides the first mechanistic connection between Gl-Rac and Gl-actin in Giardia. Native gels and overlay assays indicate intermediate proteins are required to support the interaction between Gl-14-3-3 and Gl-actin, suggesting that Gl-14-3-3 is regulating multiple Gl-actin complexes.
Collapse
|
132
|
Schoppmeyer R, Zhao R, Cheng H, Hamed M, Liu C, Zhou X, Schwarz EC, Zhou Y, Knörck A, Schwär G, Ji S, Liu L, Long J, Helms V, Hoth M, Yu X, Qu B. Human profilin 1 is a negative regulator of CTL mediated cell-killing and migration. Eur J Immunol 2017; 47:1562-1572. [PMID: 28688208 DOI: 10.1002/eji.201747124] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 05/12/2017] [Accepted: 07/05/2017] [Indexed: 12/30/2022]
Abstract
The actin-binding protein profilin1 (PFN1) plays a central role in actin dynamics, which is essential for cytotoxic T lymphocyte (CTL) functions. The functional role of PFN1 in CTLs, however still remains elusive. Here, we identify PFN1 as the only member of the profilin family expressed in primary human CD8+ T cells. Using in vitro assays, we find that PFN1 is a negative regulator of CTL-mediated elimination of target cells. Furthermore, PFN1 is involved in activation-induced lytic granule (LG) release, CTL migration and modulation of actin structures at the immunological synapse (IS). During CTL migration, PFN1 modulates the velocity, protrusion formation patterns and protrusion sustainability. In contrast, PFN1 does not significantly affect migration persistence and the rates of protrusion emergence and retraction. Under in vitro conditions mimicking a tumor microenvironment, we show that PFN1 downregulation promotes CTL invasion into a 3D matrix, without affecting the viability of CTLs in a hydrogen peroxide-enriched microenvironment. Highlighting its potential relevance in cancer, we find that in pancreatic cancer patients, PFN1 expression is substantially decreased in peripheral CD8+ T cells. Taken together, we conclude that PFN1 is a negative regulator for CTL-mediated cytotoxicity and may have an impact on CTL functionality in a tumor-related context.
Collapse
Affiliation(s)
- Rouven Schoppmeyer
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - He Cheng
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, P.R. China
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Mohamed Hamed
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Centre, Rostock, Germany
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, P.R. China
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Xiao Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Yan Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Arne Knörck
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Gertrud Schwär
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Shunrong Ji
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, P.R. China
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, P.R. China
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, P.R. China
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, P.R. China
- Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
133
|
Lappalainen P. Actin-binding proteins: the long road to understanding the dynamic landscape of cellular actin networks. Mol Biol Cell 2017; 27:2519-22. [PMID: 27528696 PMCID: PMC4985253 DOI: 10.1091/mbc.e15-10-0728] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/14/2016] [Indexed: 11/17/2022] Open
Abstract
The actin cytoskeleton supports a vast number of cellular processes in nonmuscle cells. It is well established that the organization and dynamics of the actin cytoskeleton are controlled by a large array of actin-binding proteins. However, it was only 40 years ago that the first nonmuscle actin-binding protein, filamin, was identified and characterized. Filamin was shown to bind and cross-link actin filaments into higher-order structures and contribute to phagocytosis in macrophages. Subsequently many other nonmuscle actin-binding proteins were identified and characterized. These proteins regulate almost all steps of the actin filament assembly and disassembly cycles, as well as the arrangement of actin filaments into diverse three-dimensional structures. Although the individual biochemical activities of most actin-regulatory proteins are relatively well understood, knowledge of how these proteins function together in a common cytoplasm to control actin dynamics and architecture is only beginning to emerge. Furthermore, understanding how signaling pathways and mechanical cues control the activities of various actin-binding proteins in different cellular, developmental, and pathological processes will keep researchers busy for decades.
Collapse
Affiliation(s)
- Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
134
|
Shaw AE, Bamburg JR. Peptide regulation of cofilin activity in the CNS: A novel therapeutic approach for treatment of multiple neurological disorders. Pharmacol Ther 2017; 175:17-27. [PMID: 28232023 PMCID: PMC5466456 DOI: 10.1016/j.pharmthera.2017.02.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cofilin is a ubiquitous protein which cooperates with many other actin-binding proteins in regulating actin dynamics. Cofilin has essential functions in nervous system development including neuritogenesis, neurite elongation, growth cone pathfinding, dendritic spine formation, and the regulation of neurotransmission and spine function, components of synaptic plasticity essential for learning and memory. Cofilin's phosphoregulation is a downstream target of many transmembrane signaling processes, and its misregulation in neurons has been linked in rodent models to many different neurodegenerative and neurological disorders including Alzheimer disease (AD), aggression due to neonatal isolation, autism, manic/bipolar disorder, and sleep deprivation. Cognitive and behavioral deficits of these rodent models have been largely abrogated by modulation of cofilin activity using viral-mediated, genetic, and/or small molecule or peptide therapeutic approaches. Neuropathic pain in rats from sciatic nerve compression has also been reduced by modulating the cofilin pathway within neurons of the dorsal root ganglia. Neuroinflammation, which occurs following cerebral ischemia/reperfusion, but which also accompanies many other neurodegenerative syndromes, is markedly reduced by peptides targeting specific chemokine receptors, which also modulate cofilin activity. Thus, peptide therapeutics offer potential for cost-effective treatment of a wide variety of neurological disorders. Here we discuss some recent results from rodent models using therapeutic peptides with a surprising ability to cross the rodent blood brain barrier and alter cofilin activity in brain. We also offer suggestions as to how neuronal-specific cofilin regulation might be achieved.
Collapse
Affiliation(s)
- Alisa E Shaw
- Department of Biochemistry and Molecular Biology, Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523-1870, United States
| | - James R Bamburg
- Department of Biochemistry and Molecular Biology, Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523-1870, United States.
| |
Collapse
|
135
|
Abstract
Spectacular images of the process of myosin II filament formation and organization in migrating cells are unveiled by super-resolution imaging. A combination of short- and long-range interactions with actin filaments is seen to play a critical role in filament partitioning and alignment into contractile actin arcs and stress fibres.
Collapse
Affiliation(s)
- Margaret A Titus
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
136
|
Carlier MF, Shekhar S. Global treadmilling coordinates actin turnover and controls the size of actin networks. Nat Rev Mol Cell Biol 2017. [PMID: 28248322 DOI: 10.1038/nrm.(2016)172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Various cellular processes (including cell motility) are driven by the regulated, polarized assembly of actin filaments into distinct force-producing arrays of defined size and architecture. Branched, linear, contractile and cytosolic arrays coexist in vivo, and cells intricately control the number, length and assembly rate of filaments in these arrays. Recent in vitro and in vivo studies have revealed novel molecular mechanisms that regulate the number of filament barbed and pointed ends and their respective assembly and disassembly rates, thus defining classes of dynamically different filaments, which coexist in the same cell. We propose that a global treadmilling process, in which a steady-state amount of polymerizable actin monomers is established by the dynamics of each network, is responsible for defining the size and turnover of coexisting actin networks. Furthermore, signal-induced changes in the partitioning of actin to distinct arrays (mediated by RHO GTPases) result in the establishment of various steady-state concentrations of polymerizable monomers, thereby globally influencing the growth rate of actin filaments.
Collapse
Affiliation(s)
- Marie-France Carlier
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Gif-sur-Yvette, Paris 91190, France
| | - Shashank Shekhar
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Gif-sur-Yvette, Paris 91190, France
| |
Collapse
|
137
|
Abstract
Computational simulations of polymerizing actin filaments indicate that competition for a limiting pool of building blocks is not sufficient to control their length.
Collapse
Affiliation(s)
- Cristian Suarez
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Patrick M McCall
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA; James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Margaret L Gardel
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA; James Franck Institute, The University of Chicago, Chicago, IL 60637, USA; Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
138
|
eNOS S-nitrosylates β-actin on Cys374 and regulates PKC-θ at the immune synapse by impairing actin binding to profilin-1. PLoS Biol 2017; 15:e2000653. [PMID: 28394935 PMCID: PMC5386235 DOI: 10.1371/journal.pbio.2000653] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/09/2017] [Indexed: 12/24/2022] Open
Abstract
The actin cytoskeleton coordinates the organization of signaling microclusters at the immune synapse (IS); however, the mechanisms involved remain poorly understood. We show here that nitric oxide (NO) generated by endothelial nitric oxide synthase (eNOS) controls the coalescence of protein kinase C-θ (PKC-θ) at the central supramolecular activation cluster (c-SMAC) of the IS. eNOS translocated with the Golgi to the IS and partially colocalized with F-actin around the c-SMAC. This resulted in reduced actin polymerization and centripetal retrograde flow of β-actin and PKC-θ from the lamellipodium-like distal (d)-SMAC, promoting PKC-θ activation. Furthermore, eNOS-derived NO S-nitrosylated β-actin on Cys374 and impaired actin binding to profilin-1 (PFN1), as confirmed with the transnitrosylating agent S-nitroso-L-cysteine (Cys-NO). The importance of NO and the formation of PFN1-actin complexes on the regulation of PKC-θ was corroborated by overexpression of PFN1- and actin-binding defective mutants of β-actin (C374S) and PFN1 (H119E), respectively, which reduced the coalescence of PKC-θ at the c-SMAC. These findings unveil a novel NO-dependent mechanism by which the actin cytoskeleton controls the organization and activation of signaling microclusters at the IS. T cells are an essential arm of the immunity against the invasion of pathogenic agents in organisms. These specialized cells recognize foreign antigens displayed on the surface of antigen-presenting cells (APC) by means of the T cell receptor (TCR). Early signaling takes place in these cells through the specific clustering of TCRs, which trigger the recruitment of signaling molecules to the immune synapse (IS), a plasma membrane–associated intercellular domain important for T cell activation. In this location, several signaling molecules that include the protein kinase C-θ (PKC-θ) form microclusters that are translocated centripetally towards the center of the IS, following the retrograde movement of actin. In this study, we show that nitric oxide (NO) formed by endothelial nitric oxide synthase (eNOS) regulates the translocation of PKC-θ to the IS, increasing its activation. eNOS can effectively modify β-actin by S-nitrosylation on Cys374, reducing its ability to bind profilin-1 (PFN1)—a protein required for actin polymerization—polymerize and flow from the periphery to the central region of the IS. We propose that eNOS-derived NO controls actin polymerization via S-nitrosylation of actin as one of the major driving forces for the transport of PKC-θ towards the central area of the IS, which is essential for T cell activation.
Collapse
|
139
|
Nejedla M, Li Z, Masser AE, Biancospino M, Spiess M, Mackowiak SD, Friedländer MR, Karlsson R. A Fluorophore Fusion Construct of Human Profilin I with Non-Compromised Poly(L-Proline) Binding Capacity Suitable for Imaging. J Mol Biol 2017; 429:964-976. [DOI: 10.1016/j.jmb.2017.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/06/2016] [Accepted: 01/03/2017] [Indexed: 10/24/2022]
|
140
|
Burke TA, Harker AJ, Dominguez R, Kovar DR. The bacterial virulence factors VopL and VopF nucleate actin from the pointed end. J Cell Biol 2017; 216:1267-1276. [PMID: 28363971 PMCID: PMC5412564 DOI: 10.1083/jcb.201608104] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 11/22/2022] Open
Abstract
How the bacterial virulence factors VopL/F from Vibrio catalyze actin nucleation is unclear. Using multicolor TIRF microscopy imaging, Burke et al. find that VopL and VopF stimulate actin assembly via identical pointed-end nucleation mechanisms. VopL and VopF (VopL/F) are tandem WH2-domain actin assembly factors used by infectious Vibrio species to induce actin assembly in host cells. There is disagreement about the filament assembly mechanism of VopL/F, including whether they associate with the filament barbed or pointed end. Here, we used multicolor total internal reflection fluorescence microscopy to directly observe actin assembly with fluorescently labeled VopL/F. In actin monomer assembly reactions, VopL/F exclusively nucleate actin filament assemblies, remaining only briefly associated with the pointed end. VopL/F do not associate with the ends of preassembled filaments. In assembly reactions with saturating profilin, ∼85% of VopL/F molecules also promote nucleation from the pointed end, whereas a smaller fraction (<15%) associate for ∼25 s with the barbed end of preassembled filaments, inhibiting their elongation. We conclude that VopL/F function primarily as actin nucleation factors that remain briefly (∼100 s) associated with the pointed end.
Collapse
Affiliation(s)
- Thomas A Burke
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Alyssa J Harker
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637 .,Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
141
|
Dimchev G, Steffen A, Kage F, Dimchev V, Pernier J, Carlier MF, Rottner K. Efficiency of lamellipodia protrusion is determined by the extent of cytosolic actin assembly. Mol Biol Cell 2017; 28:1311-1325. [PMID: 28331069 PMCID: PMC5426846 DOI: 10.1091/mbc.e16-05-0334] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 02/10/2017] [Accepted: 03/17/2017] [Indexed: 11/26/2022] Open
Abstract
Lamellipodia protrusion requires actin network formation driven by the Arp2/3 complex and its upstream regulators WAVE complex and Rac. Actin assembly factors of the formin and Ena/VASP families can influence protrusion, in particular by maintaining a balance between lamellipodial and cytosolic actin filament assembly. Cell migration and cell–cell communication involve the protrusion of actin-rich cell surface projections such as lamellipodia and filopodia. Lamellipodia are networks of actin filaments generated and turned over by filament branching through the Arp2/3 complex. Inhibition of branching is commonly agreed to eliminate formation and maintenance of lamellipodial actin networks, but the regulation of nucleation or elongation of Arp2/3-independent filament populations within the network by, for example, formins or Ena/VASP family members and its influence on the effectiveness of protrusion have been unclear. Here we analyzed the effects of a set of distinct formin fragments and VASP on site-specific, lamellipodial versus cytosolic actin assembly and resulting consequences on protrusion. Surprisingly, expression of formin variants but not VASP reduced lamellipodial protrusion in B16-F1 cells, albeit to variable extents. The rates of actin network polymerization followed a similar trend. Unexpectedly, the degree of inhibition of both parameters depended on the extent of cytosolic but not lamellipodial actin assembly. Indeed, excess cytosolic actin assembly prevented actin monomer from rapid translocation to and efficient incorporation into lamellipodia. Thus, as opposed to sole regulation by actin polymerases operating at their tips, the protrusion efficiency of lamellipodia is determined by a finely tuned balance between lamellipodial and cytosolic actin assembly.
Collapse
Affiliation(s)
- Georgi Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Vanessa Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Julien Pernier
- Cytoskeleton Dynamics and Motility, Centre National de la Recherche Scientifique, 91198 Gif-sur-Yvette, France
| | - Marie-France Carlier
- Cytoskeleton Dynamics and Motility, Centre National de la Recherche Scientifique, 91198 Gif-sur-Yvette, France
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany .,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
142
|
Christensen JR, Hocky GM, Homa KE, Morganthaler AN, Hitchcock-DeGregori SE, Voth GA, Kovar DR. Competition between Tropomyosin, Fimbrin, and ADF/Cofilin drives their sorting to distinct actin filament networks. eLife 2017; 6. [PMID: 28282023 PMCID: PMC5404920 DOI: 10.7554/elife.23152] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/09/2017] [Indexed: 12/15/2022] Open
Abstract
The fission yeast actin cytoskeleton is an ideal, simplified system to investigate fundamental mechanisms behind cellular self-organization. By focusing on the stabilizing protein tropomyosin Cdc8, bundling protein fimbrin Fim1, and severing protein coffin Adf1, we examined how their pairwise and collective interactions with actin filaments regulate their activity and segregation to functionally diverse F-actin networks. Utilizing multi-color TIRF microscopy of in vitro reconstituted F-actin networks, we observed and characterized two distinct Cdc8 cables loading and spreading cooperatively on individual actin filaments. Furthermore, Cdc8, Fim1, and Adf1 all compete for association with F-actin by different mechanisms, and their cooperative association with actin filaments affects their ability to compete. Finally, competition between Fim1 and Adf1 for F-actin synergizes their activities, promoting rapid displacement of Cdc8 from a dense F-actin network. Our findings reveal that competitive and cooperative interactions between actin binding proteins help define their associations with different F-actin networks. DOI:http://dx.doi.org/10.7554/eLife.23152.001 Cells use a protein called actin to provide shape, to generate the forces needed for cells to divide, and for many other essential processes. Inside a cell, individual actin proteins join up to form long filaments. These actin filaments are organized in different ways to make networks that have distinct properties, each tailored for a specific process. For instance, bundles of straight actin filaments help a cell to divide, whereas a network of branched actin filaments allows cells to move. The different proteins that bind to actin filaments influence how quickly actin filaments are assembled and organized into networks. Therefore, many of the properties of an actin filament network are due to the actin binding proteins that are associated with it. Two actin binding proteins called fimbrin and cofilin associate with a type of actin filament network known as the actin patch. A third actin binding protein called tropomyosin associates with a different network that forms a ring. It is not known how particular actin binding proteins choose to associate with one actin network instead of another. Christensen et al. used a fluorescence microscopy technique to study how fimbrin, cofilin and tropomyosin associate with different actin networks in a single-celled organism called fission yeast. This technique involved incubating actin and actin binding proteins together in a microscope chamber. The experiments show that some actin binding proteins, like tropomyosin, cooperate to bind to actin. Individual tropomyosin molecules find it difficult to bind actin filaments on their own, but once one tropomyosin molecule is attached to the filament, others rapidly join to coat the filament. On the other hand, some actin-binding proteins compete for binding to filaments. For example, the binding of fimbrin to actin filaments causes tropomyosin to be removed from the actin network. Further experiments revealed that fimbrin and cofilin work with each other to rapidly generate a dense actin network and displace tropomyosin. Together, the findings of Christensen et al. suggest that competitions between actin binding proteins determine which actin binding proteins are associated with an actin network. The next challenge is to understand how the most competitive actin-binding proteins are kept off actin networks where they do not belong. Further studies will shed light on how these interactions cause large changes in how the cell is organized. DOI:http://dx.doi.org/10.7554/eLife.23152.002
Collapse
Affiliation(s)
- Jenna R Christensen
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Glen M Hocky
- Department of Chemistry, The University of Chicago, Chicago, United States.,James Franck Institute, The University of Chicago, Chicago, United States
| | - Kaitlin E Homa
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Alisha N Morganthaler
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Sarah E Hitchcock-DeGregori
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, United States
| | - Gregory A Voth
- Department of Chemistry, The University of Chicago, Chicago, United States.,James Franck Institute, The University of Chicago, Chicago, United States.,Computation Institute, The University of Chicago, Chicago, United States.,Institute for Biophysical Dynamics, The University of Chicago, Chicago, United States
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States.,Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, United States
| |
Collapse
|
143
|
Thieleke-Matos C, Osório DS, Carvalho AX, Morais-de-Sá E. Emerging Mechanisms and Roles for Asymmetric Cytokinesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:297-345. [PMID: 28526136 DOI: 10.1016/bs.ircmb.2017.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytokinesis completes cell division by physically separating the contents of the mother cell between the two daughter cells. This event requires the highly coordinated reorganization of the cytoskeleton within a precise window of time to ensure faithful genomic segregation. In addition, recent progress in the field highlighted the importance of cytokinesis in providing particularly important cues in the context of multicellular tissues. The organization of the cytokinetic machinery and the asymmetric localization or inheritance of the midbody remnants is critical to define the spatial distribution of mechanical and biochemical signals. After a brief overview of the conserved steps of animal cytokinesis, we review the mechanisms controlling polarized cytokinesis focusing on the challenges of epithelial cytokinesis. Finally, we discuss the significance of these asymmetries in defining embryonic body axes, determining cell fate, and ensuring the correct propagation of epithelial organization during proliferation.
Collapse
Affiliation(s)
- C Thieleke-Matos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - D S Osório
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A X Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cytoskeletal Dynamics, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - E Morais-de-Sá
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Cell Division and Genomic stability, IBMC, Instituto de Biologia Molecular e Celular, and i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
144
|
Carlier MF, Shekhar S. Global treadmilling coordinates actin turnover and controls the size of actin networks. Nat Rev Mol Cell Biol 2017; 18:389-401. [DOI: 10.1038/nrm.2016.172] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
145
|
Actin dynamics and competition for myosin monomer govern the sequential amplification of myosin filaments. Nat Cell Biol 2017; 19:85-93. [PMID: 28114272 PMCID: PMC5308804 DOI: 10.1038/ncb3463] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022]
Abstract
The cellular mechanisms governing non-muscle myosin II (NM2) filament assembly are largely unknown. Using EGFP-NM2A knock-in fibroblasts and multiple super-resolution imaging modalities, we characterized and quantified the sequential amplification of NM2 filaments within lamellae, wherein filaments emanating from single nucleation events continuously partition, forming filament clusters that populate large-scale actomyosin structures deeper in the cell. Individual partitioning events coincide spatially and temporally with the movements of diverging actin fibres, suppression of which inhibits partitioning. These and other data indicate that NM2A filaments are partitioned by the dynamic movements of actin fibres to which they are bound. Finally, we showed that partition frequency and filament growth rate in the lamella depend on MLCK, and that MLCK is competing with centrally active ROCK for a limiting pool of monomer with which to drive lamellar filament assembly. Together, our results provide new insights into the mechanism and spatio-temporal regulation of NM2 filament assembly in cells.
Collapse
|
146
|
Sedzinski J, Hannezo E, Tu F, Biro M, Wallingford JB. RhoA regulates actin network dynamics during apical surface emergence in multiciliated epithelial cells. J Cell Sci 2017; 130:420-428. [PMID: 28089989 PMCID: PMC5278671 DOI: 10.1242/jcs.194704] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/10/2016] [Indexed: 12/18/2022] Open
Abstract
Homeostatic replacement of epithelial cells from basal precursors is a multistep process involving progenitor cell specification, radial intercalation and, finally, apical surface emergence. Recent data demonstrate that actin-based pushing under the control of the formin protein Fmn1 drives apical emergence in nascent multiciliated epithelial cells (MCCs), but little else is known about this actin network or the control of Fmn1. Here, we explore the role of the small GTPase RhoA in MCC apical emergence. Disruption of RhoA function reduced the rate of apical surface expansion and decreased the final size of the apical domain. Analysis of cell shapes suggests that RhoA alters the balance of forces exerted on the MCC apical surface. Finally, quantitative time-lapse imaging and fluorescence recovery after photobleaching studies argue that RhoA works in concert with Fmn1 to control assembly of the specialized apical actin network in MCCs. These data provide new molecular insights into epithelial apical surface assembly and could also shed light on mechanisms of apical lumen formation.
Collapse
Affiliation(s)
- Jakub Sedzinski
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Edouard Hannezo
- Cavendish Laboratory, Department of Physics, J.J. Thomson Avenue, University of Cambridge, Cambridge CB3 0HE, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Fan Tu
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Maté Biro
- Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag 6, Newtown, New South Wales 2042, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
147
|
Abstract
Actin functions in a multitude of cellular processes owing to its ability to polymerize into filaments, which can be further organized into higher-order structures by an array of actin-binding and regulatory proteins. Therefore, research on actin and actin-related functions relies on the visualization of actin structures without interfering with the cycles of actin polymerization and depolymerization that underlie cellular actin dynamics. In this Cell Science at a Glance and the accompanying poster, we briefly evaluate the different techniques and approaches currently applied to analyze and visualize cellular actin structures, including in the nuclear compartment. Referring to the gold standard F-actin marker phalloidin to stain actin in fixed samples and tissues, we highlight methods for visualization of actin in living cells, which mostly apply the principle of genetically fusing fluorescent proteins to different actin-binding domains, such as LifeAct, utrophin and F-tractin, as well as anti-actin-nanobody technology. In addition, the compound SiR-actin and the expression of GFP-actin are also applicable for various types of live-cell analyses. Overall, the visualization of actin within a physiological context requires a careful choice of method, as well as a tight control of the amount or the expression level of a given detection probe in order to minimize its influence on endogenous actin dynamics.
Collapse
Affiliation(s)
- Michael Melak
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), University of Marburg, Karl-von-Frisch-Straße 1, Marburg 35043, Germany
| | - Matthias Plessner
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), University of Marburg, Karl-von-Frisch-Straße 1, Marburg 35043, Germany
| | - Robert Grosse
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), University of Marburg, Karl-von-Frisch-Straße 1, Marburg 35043, Germany
| |
Collapse
|
148
|
Wales P, Schuberth CE, Aufschnaiter R, Fels J, García-Aguilar I, Janning A, Dlugos CP, Schäfer-Herte M, Klingner C, Wälte M, Kuhlmann J, Menis E, Hockaday Kang L, Maier KC, Hou W, Russo A, Higgs HN, Pavenstädt H, Vogl T, Roth J, Qualmann B, Kessels MM, Martin DE, Mulder B, Wedlich-Söldner R. Calcium-mediated actin reset (CaAR) mediates acute cell adaptations. eLife 2016; 5. [PMID: 27919320 PMCID: PMC5140269 DOI: 10.7554/elife.19850] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/14/2016] [Indexed: 12/24/2022] Open
Abstract
Actin has well established functions in cellular morphogenesis. However, it is not well understood how the various actin assemblies in a cell are kept in a dynamic equilibrium, in particular when cells have to respond to acute signals. Here, we characterize a rapid and transient actin reset in response to increased intracellular calcium levels. Within seconds of calcium influx, the formin INF2 stimulates filament polymerization at the endoplasmic reticulum (ER), while cortical actin is disassembled. The reaction is then reversed within a few minutes. This Calcium-mediated actin reset (CaAR) occurs in a wide range of mammalian cell types and in response to many physiological cues. CaAR leads to transient immobilization of organelles, drives reorganization of actin during cell cortex repair, cell spreading and wound healing, and induces long-lasting changes in gene expression. Our findings suggest that CaAR acts as fundamental facilitator of cellular adaptations in response to acute signals and stress. DOI:http://dx.doi.org/10.7554/eLife.19850.001 Our skeleton plays a vital role in giving shape and structure to our body, it also allows us to make coordinated movements. Similarly, each cell contains a microscopic network of structures and supports called the cytoskeleton that helps cells to adopt specific shapes and is crucial for them to move around. Unlike our skeleton, which is relatively unchanging, the cytoskeleton of each cell constantly changes and adapts to the specific needs of the cell. One part of the cytoskeleton is a dense, flexible meshwork of fibers called the cortex that lies just beneath the surface of the cell. The cortex is constructed using a protein called actin, and many of these proteins join together to form each fiber. When cells need to adapt rapidly to an injury or other sudden changes in their environment they activate a so-called stress response. This response often begins with a rapid increase in the amount of calcium ions inside a cell, which can then trigger changes in actin organization. However, it is not clear how cells under stress are able to globally remodel their actin cytoskeleton without compromising stability and integrity of the cortex. Wales, Schuberth, Aufschnaiter et al. used a range of mammalian cells to investigate how actin responds to stress signals. All cells responded to the resulting influx of calcium ions by deconstructing large parts of the actin cortex and simultaneously forming actin filaments near the center of the cell. Wales, Schuberth, Aufschnaiter et al. termed this response calcium-mediated actin reset (CaAR), as it lasted for only a few minutes before the actin cortex reformed. The experiments show that a protein called INF2 controls CaAR by rapidly removing actin from the cortex and forming new filaments near a cell compartment called the endoplasmic reticulum. CaAR allows cells to rapidly and drastically alter the cortex in response to stress. The experiments also show that this sudden shift in actin can change the activity of certain genes, leading to longer-term effects on the cell. The findings of Wales, Schuberth, Aufschnaiter et al. suggest that calcium ions globally regulate the actin cytoskeleton and hence cell shape and movement under stress. This could be relevant for many important processes and conditions such as wound healing, inflammation and cancer. A future challenge will be to understand the role of CaAR in these processes. DOI:http://dx.doi.org/10.7554/eLife.19850.002
Collapse
Affiliation(s)
- Pauline Wales
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Christian E Schuberth
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Roland Aufschnaiter
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Johannes Fels
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | | | - Annette Janning
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Christopher P Dlugos
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany.,Medical Clinic D, University Clinic of Muenster, Muenster, Germany
| | - Marco Schäfer-Herte
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Christoph Klingner
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany.,AG Molecular Mechanotransduction, Max Planck Institute of Biochemistry, Munich, Germany
| | - Mike Wälte
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Julian Kuhlmann
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Ekaterina Menis
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Laura Hockaday Kang
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| | - Kerstin C Maier
- Department of Biochemistry, University of Munich, Munich, Germany
| | - Wenya Hou
- Institute of Biochemistry I, Friedrich Schiller University Jena, Jena, Germany
| | - Antonella Russo
- Institute of Immunology, University of Münster, Münster, Germany
| | - Henry N Higgs
- Department of Biochemistry, Dartmouth Medical School, Hanover, United States
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Friedrich Schiller University Jena, Jena, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Friedrich Schiller University Jena, Jena, Germany
| | - Dietmar E Martin
- Department of Biochemistry, University of Munich, Munich, Germany
| | - Bela Mulder
- Theory of Biological Matter, FOM Institute AMOLF, Amsterdam, Netherlands
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, University of Muenster, Muenster, Germany.,Cells-In-Motion Cluster of Excellence (EXC1003 - CiM), University of Münster, Muenster, Germany
| |
Collapse
|
149
|
Alioto SL, Garabedian MV, Bellavance DR, Goode BL. Tropomyosin and Profilin Cooperate to Promote Formin-Mediated Actin Nucleation and Drive Yeast Actin Cable Assembly. Curr Biol 2016; 26:3230-3237. [PMID: 27866892 DOI: 10.1016/j.cub.2016.09.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/01/2016] [Accepted: 09/26/2016] [Indexed: 01/06/2023]
Abstract
Tropomyosins comprise a large family of actin-binding proteins with critical roles in diverse actin-based processes [1], but our understanding of how they mechanistically contribute to actin filament dynamics has been limited. We addressed this question in S. cerevisiae, where tropomyosins (Tpm1 and Tpm2), profilin (Pfy1), and formins (Bni1 and Bnr1) are required for the assembly of an array of actin cables that facilitate polarized vesicle delivery and daughter cell growth. Formins drive cable formation by promoting actin nucleation and by accelerating actin filament elongation together with profilin [2]. In contrast, how tropomyosins contribute mechanistically to cable formation has been unclear, but genetic studies demonstrate that Tpm1 plays a more important role than Tpm2 [3, 4]. Here, we found that loss of TPM1 in strains lacking BNR1, but not BNI1, leads to severe defects in cable formation, polarized secretion, and cell growth, suggesting that TPM1 function is required for proper Bni1-mediated cable assembly. Furthermore, in vitro total internal reflection fluorescence (TIRF) microscopy demonstrated that Tpm1 strongly enhances Bni1-mediated, but not Bnr1-mediated, actin nucleation without affecting filament elongation rate, whereas Tpm2 has no effects on Bni1 or Bnr1. Tpm1 stimulation of Bni1-mediated nucleation also requires profilin and its interactions with both G-actin and formins. Together, these results demonstrate that yeast Tpm1 works in concert with profilin to promote formin-dependent nucleation of actin cables, thus expanding our understanding of how specific tropomyosin isoforms influence actin dynamics.
Collapse
Affiliation(s)
- Salvatore L Alioto
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA
| | - Mikael V Garabedian
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA
| | - Danielle R Bellavance
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA
| | - Bruce L Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
150
|
Kapustina M, Read TA, Vitriol EA. Simultaneous quantification of actin monomer and filament dynamics with modeling-assisted analysis of photoactivation. J Cell Sci 2016; 129:4633-4643. [PMID: 27831495 PMCID: PMC5201019 DOI: 10.1242/jcs.194670] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/02/2016] [Indexed: 01/30/2023] Open
Abstract
Photoactivation allows one to pulse-label molecules and obtain quantitative data about their behavior. We have devised a new modeling-based analysis for photoactivatable actin experiments that simultaneously measures properties of monomeric and filamentous actin in a three-dimensional cellular environment. We use this method to determine differences in the dynamic behavior of β- and γ-actin isoforms, showing that both inhabit filaments that depolymerize at equal rates but that β-actin exists in a higher monomer-to-filament ratio. We also demonstrate that cofilin (cofilin 1) equally accelerates depolymerization of filaments made from both isoforms, but is only required to maintain the β-actin monomer pool. Finally, we used modeling-based analysis to assess actin dynamics in axon-like projections of differentiating neuroblastoma cells, showing that the actin monomer concentration is significantly depleted as the axon develops. Importantly, these results would not have been obtained using traditional half-time analysis. Given that parameters of the publicly available modeling platform can be adjusted to suit the experimental system of the user, this method can easily be used to quantify actin dynamics in many different cell types and subcellular compartments.
Collapse
Affiliation(s)
- Maryna Kapustina
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tracy-Ann Read
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | - Eric A Vitriol
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|